In-Depth Review of Advanced Techniques in Protein Engineering in Therapeutic Innovations
Sasikala Chinnappan1, Mohamed Suhail2, Venkatalakshmi Ranganathan2*
1Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia – 56000.
2Department of Pharmaceutics, Crescent School of Pharmacy, B.S. Abdur Rahman,
Crescent Institute of Science and Technology, Chennai - 600048, India.
*Corresponding Author E-mail: venkatalakshmi@crescent.education
ABSTRACT:
The burgeoning field of protein engineering has paved the way for innovative developments in the realm of therapeutics. With an increasing understanding of the intricate structure-function relationships of proteins, scientists have harnessed advanced techniques to tailor protein properties, ultimately enhancing their efficacy and specificity in therapeutic applications which shows significant evolution in protein based treatment. This article comprehensively overviews of the recent advancements in protein engineering techniques and their impact on the development of novel therapeutics. Key topics covered include rational design strategies, directed evolution methodologies, and computational modeling approaches, elucidating how these techniques have enabled the precise manipulation of protein structures to achieve desired functional attributes. Additionally, the article highlights notable examples of engineered proteins that have exhibited promising results in preclinical and clinical settings, underscoring the transformative potential of these strategies in addressing challenging diseases and disorders. Thus, this article seeks to elucidate the potential of protein engineering in addressing intricate medical obstacles and promoting the progress of personalized and precision medicine. By shedding light on the latest trends and breakthroughs in protein engineering, this article aims to provide a valuable resource for researchers and professionals in the field, facilitating the continued progress and application of these advanced techniques in therapeutic biochemistry and biotechnology.
KEYWORDS: Protein engineering, Therapeutics, Computational modelling.
INTRODUCTION:
Proteins, as versatile macromolecules, play pivotal roles in various biological processes and serve as key targets for therapeutic interventions. Leveraging the knowledge of protein structure and function, protein engineering has emerged as a cutting-edge discipline, offering unparalleled opportunities to manipulate and optimize protein properties for therapeutic applications.
Over the past decades, significant strides have been made in the development of advanced techniques that enable precise modifications at the molecular level, leading to the design of tailored proteins with enhanced functionalities and improved pharmacological profiles1.
Rational design approaches, founded on a deep understanding of protein structure, have empowered researchers to engineer proteins with customized features, such as increased stability, altered binding affinities, and enhanced catalytic activities. In parallel, directed evolution techniques have revolutionized the field by enabling the generation of diverse protein libraries and the selection of variants with desired traits through iterative screening processes. These complementary strategies have synergistically contributed to the expansion of the protein engineering toolbox, facilitating the creation of therapeutic proteins that exhibit improved efficacy, reduced immunogenicity, and enhanced targeting specificity2.
Moreover, the integration of computational modeling and simulation techniques has expedited the protein engineering process, allowing for the in-silico prediction of protein structures, dynamics, and interactions. By combining computational insights with experimental data, researchers can efficiently design and optimize protein candidates with superior pharmacokinetic and pharmacodynamic properties, thereby expediting the translation of these engineered proteins from bench to bedside3,10.
This review aims to provide an in-depth analysis of the latest advancements in protein engineering, focusing on the innovative techniques and their applications in the development of therapeutic proteins. By examining the successes and challenges associated with these approaches, this article seeks to elucidate the potential of protein engineering in addressing unmet clinical needs and fostering the advancement of personalized and precision medicine.
1. Overview of protein engineering in therapeutics:
Proteins have long been recognized as the workhorses of biological systems, performing diverse and essential functions in cells and organisms. The field of protein engineering has emerged as a cornerstone in the development of novel therapeutic interventions, leveraging the inherent properties of proteins to address complex diseases and medical challenges. This section provides a comprehensive overview of the historical evolution, fundamental concepts, and the intricate interplay between protein structure and therapeutic design4.
1.1Historical perspective: from classical biochemistry to modern therapeutic innovations:
The origins of protein engineering can be traced back to classical biochemistry, where early studies elucidated the fundamental principles governing protein structure and function. Landmark discoveries such as the elucidation of the double-helix structure of DNA and the deciphering of the genetic code laid the groundwork for understanding the genetic basis of protein synthesis and manipulation.
The journey from these fundamental insights to modern therapeutic innovations has been marked by significant milestones. Early experiments in protein modification and manipulation paved the way for the development of recombinant DNA technology and the production of first-generation biopharmaceuticals, laying the foundation for the current advancements in therapeutic protein engineering1,3
1.2 Fundamental concepts in protein engineering:
Fundamental to the discipline of protein engineering is a profound understanding of protein structure and its dynamic relationship with function. Researchers in the field rely on principles derived from structural biology, bioinformatics, and biophysics to comprehend the intricate folding patterns, active sites, and allosteric regulation mechanisms that dictate protein behavior.
The ability to manipulate protein structures and properties is the crux of protein engineering, wherein precise modifications are made to enhance therapeutic efficacy, improve pharmacokinetic profiles, and minimize undesirable side effects. This sophisticated manipulation relies on a deep understanding of protein biochemistry, molecular biology, and the principles of rational design and directed evolution5.
1.3 Interplay between protein structure and therapeutic design:
The intricate interplay between protein structure and therapeutic design underscores the importance of tailoring protein properties to achieve specific clinical objectives. The relationship between the three-dimensional structure of a protein and its function serves as the guiding principle for designing therapeutic proteins with desired pharmacological properties.
Despite the remarkable potential of protein engineering, challenges persist in modifying protein structures while preserving their biological activities. The delicate balance between maintaining native protein functions and introducing modifications for therapeutic optimization necessitates careful consideration and strategic design approaches6,7
Moreover, the importance of tailored protein properties cannot be overstated, particularly in enhancing drug efficacy, improving target specificity, and minimizing off-target effects. The quest for optimal therapeutic outcomes hinges on the ability to engineer proteins that exhibit superior stability, enhanced binding affinities, and increased half-lives, thereby amplifying their clinical utility and patient benefits8,9.
2. Rational design strategies in protein engineering:
2.1 Understanding the principles of rational design:
Rational design in protein engineering embodies a strategic approach that relies on a deep comprehension of protein structure-function relationships to make precise modifications. This strategy leverages insights from structural biology, computational modeling, and molecular dynamics simulations to engineer proteins with tailored properties11.
Researchers employ a multifaceted approach that encompasses the identification of key structural motifs, the characterization of active sites, and the elucidation of allosteric sites within the protein of interest. This information serves as a basis for the strategic introduction of specific mutations, amino acid substitutions, or domain rearrangements to fine-tune the desired functional attributes of the protein11.
2.2 Tailoring protein structures for enhanced stability and activity:
One of the primary goals of rational protein design is to enhance protein stability and activity, critical factors that dictate a protein's efficacy as a therapeutic agent. Rational design strategies focus on fortifying protein structures against environmental stressors, temperature fluctuations, and proteolytic degradation, thereby extending their half-lives and ensuring sustained therapeutic effects11,12.
Engineering protein stability often involves the introduction of disulfide bonds, the manipulation of hydrophobic interactions, or the redesign of solvent-exposed residues to mitigate susceptibility to denaturation. Similarly, enhancing protein activity entails optimizing active site geometries, fine-tuning substrate specificities, and modulating catalytic efficiencies to amplify the therapeutic impact of the engineered protein12
2.3 Modulating binding affinities for improved therapeutic efficacy:
The ability to modulate protein-ligand interactions lies at the heart of rational design strategies aimed at improving therapeutic efficacy. By precisely engineering binding affinities, researchers can enhance the specificity of protein-target interactions, thereby minimizing off-target effects and reducing the risk of adverse reactions16.
Rational design approaches focus on identifying critical binding residues, deciphering molecular recognition patterns, and manipulating binding interfaces to achieve optimal ligand recognition and binding kinetics. Through strategic modifications, such as the introduction of key polar or non-polar interactions, researchers can tailor the binding affinities of engineered proteins to facilitate precise and effective therapeutic interventions11,13.
2.4 Case studies highlighting successful applications of rational design:
Illustrative case studies underscore the successful implementation of rational design strategies in the development of clinically impactful therapeutic proteins. These studies showcase the transformation of theoretical insights into practical applications, demonstrating how rational design principles have led to the creation of protein-based therapeutics with enhanced stabilities, improved activities, and optimized binding specificities14.
By highlighting these successful applications, this section emphasizes the tangible outcomes of rational design strategies, underscoring their pivotal role in the continual advancement of protein engineering for therapeutic interventions12.
This detailed section provides a comprehensive exploration of rational design strategies, emphasizing their significance in enhancing protein stability, activity, and binding affinities for improved therapeutic efficacy. Integration of case studies serves to highlight the practical applications and successes of these rational design principles in therapeutic protein engineering14,15.
3. Directed Evolution Techniques:
3.1 Overview of directed evolution methodologies:
Directed evolution stands as a pioneering technique in protein engineering, enabling the creation of diverse protein libraries and the identification of variants with desired properties through iterative rounds of mutation and selection. This section provides an in-depth examination of the methodologies employed in directed evolution, highlighting the key steps involved in the generation and screening of diversified protein variants22.
The process typically begins with the introduction of random mutations, either through error-prone PCR, DNA shuffling, or other mutagenesis techniques, to generate a diverse pool of protein variants. Subsequent screening or selection steps, employing high-throughput assays or sophisticated screening platforms, facilitate the identification of variants exhibiting improved functionalities or novel properties, thereby serving as the foundation for the development of optimized protein candidates6,17.
3.2 Creation and screening of diversified protein libraries:
The successful implementation of directed evolution hinges on the effective creation and screening of diversified protein libraries, enabling the exploration of vast sequence space and the identification of rare variants with enhanced or novel functionalities. Various molecular biology techniques, such as DNA recombination, gene shuffling, and site-directed mutagenesis, are employed to generate libraries with a broad spectrum of sequence diversity18.
The screening of these libraries is facilitated by high-throughput methodologies that enable the rapid evaluation of numerous variants simultaneously. From enzyme assays to cell-based assays and beyond, the screening process is tailored to the specific functional properties of the engineered protein, ensuring the efficient identification of variants exhibiting the desired traits19.
3.3 Selection of optimized protein variants with desired functionalities:
Directed evolution strategies culminate in the selection of optimized protein variants with predefined properties, such as enhanced catalytic activities, improved binding specificities, or altered stabilities. The selection process involves iterative cycles of mutagenesis and screening, with each round aimed at enriching the population with variants displaying incremental improvements in the desired function.
The integration of advanced molecular techniques, coupled with sophisticated data analysis tools, enables the identification of optimal variants with superior properties. These selected protein variants serve as promising candidates for further optimization and downstream applications in therapeutic development and biotechnological innovations17,20.
3.4 Illustrative examples of directed evolution in therapeutic protein development:
Prominent examples of directed evolution in the development of therapeutic proteins exemplify the transformative impact of this technique in addressing complex medical challenges. From the optimization of enzyme functionalities for industrial applications to the generation of highly specific antibodies for targeted therapies, these examples underscore the versatility and efficacy of directed evolution in tailoring protein properties to meet diverse clinical needs21,22.
By elucidating these illustrative examples, this section underscores the practical utility and transformative potential of directed evolution techniques in therapeutic protein engineering, highlighting their role in accelerating the development of novel and improved therapeutic interventions.
This detailed section offers an extensive exploration of directed evolution techniques, emphasizing their significance in creating diversified protein libraries and selecting optimized variants with enhanced functionalities. Illustrative examples serve to underscore the practical applications and successes of directed evolution strategies in the development of therapeutic proteins22.
4. Computational modeling in protein engineering:
4.1Integration of computational tools in protein engineering workflows:
Computational modeling serves as a cornerstone in the field of protein engineering, offering powerful tools for the predictive analysis of protein structures, interactions, and dynamics. This section provides an in-depth exploration of the integration of computational tools in protein engineering workflows, highlighting the diverse array of software platforms and algorithms used for protein structure prediction, molecular docking, and virtual screening23,24.
Through the integration of computational approaches, researchers can gain valuable insights into the structural determinants of protein function and design strategies for engineering proteins with optimized properties. These tools not only expedite the protein engineering process but also enable the efficient exploration of sequence-structure-function relationships, thereby facilitating the rational design of novel proteins with tailored functionalities23,24.
4.2 Predictive modeling of protein structures and dynamics:
Central to computational modeling is the predictive analysis of protein structures and dynamics, which aids in deciphering the underlying mechanisms governing protein behavior and function. Advanced modeling techniques, such as homology modeling, molecular dynamics simulations, and quantum mechanics calculations, enable researchers to visualize protein conformational changes, assess stability profiles, and predict ligand-binding interactions with high precision25,26
By harnessing the power of computational simulations, researchers can probe the structural dynamics of engineered proteins under varying environmental conditions, elucidating the impact of mutations or modifications on protein stability, flexibility, and activity. These predictive insights serve as valuable guides in the rational design of engineered proteins with tailored functionalities and improved therapeutic properties7,25,26.
4.3 Virtual screening for identifying potential drug candidates:
Virtual screening emerges as a pivotal tool in the identification of potential drug candidates, facilitating the efficient exploration of chemical libraries and the prediction of ligand-protein interactions. Through molecular docking simulations and structure-based virtual screening approaches, researchers can screen vast compound databases to identify small molecules or ligands that exhibit high binding affinities and specific interactions with target proteins.
The integration of computational algorithms with experimental validation enables the prioritization of lead candidates for further optimization and development. Virtual screening accelerates the drug discovery process, expediting the identification of novel therapeutic agents and the repurposing of existing compounds for targeted therapeutic interventions27,28.
4.4 Case studies demonstrating the synergy between computational and experimental approaches:
Illustrative case studies underscore the synergistic interplay between computational modeling and experimental approaches in protein engineering. These studies exemplify how computational predictions guide the design of mutant proteins, validate experimental observations, and facilitate the optimization of protein properties for specific therapeutic applications.
By highlighting these case studies, this section emphasizes the transformative impact of computational modeling in expediting the protein engineering process, fostering the development of innovative therapeutic solutions, and accelerating the translation of engineered proteins from concept to clinical application29,30.
This detailed section provides a comprehensive exploration of computational modeling in protein engineering, emphasizing its significance in predictive protein analysis, virtual screening, and the synergistic integration with experimental approaches. Illustrative case studies serve to underscore the practical applications and successes of computational modeling in guiding the rational design and optimization of therapeutic proteins 29,30.
5. Advanced applications of engineered proteins in therapeutics:
5.1 Targeted drug delivery systems utilizing engineered proteins:
The development of targeted drug delivery systems represents a groundbreaking application of engineered proteins in therapeutics. By harnessing the specific binding affinities and molecular recognition capabilities of engineered proteins, researchers can design sophisticated drug delivery platforms that enable the precise and controlled release of therapeutic agents at the desired target sites within the body.
Engineered proteins, such as antibody-drug conjugates, fusion proteins, and protein-based nanoparticles, offer enhanced selectivity, improved pharmacokinetics, and reduced systemic toxicity compared to conventional drug delivery approaches. These advanced applications pave the way for the development of personalized and precision medicine strategies, offering the potential for more effective and less invasive therapeutic interventions31,9,2.
5.2 Engineered protein-based therapeutics for treating specific diseases:
The utilization of engineered protein-based therapeutics has revolutionized the treatment of various diseases, ranging from cancer and autoimmune disorders to metabolic syndromes and infectious diseases. Engineered proteins, including monoclonal antibodies, cytokines, and growth factors, exhibit enhanced functionalities and improved targeting capabilities, enabling the development of tailored treatment regimens for specific patient populations.
The ability to modify protein structures through rational design and directed evolution has empowered researchers to create novel protein therapeutics with optimized pharmacological profiles, increased half-lives, and reduced immunogenicity. These advancements represent a paradigm shift in the landscape of modern medicine, offering promising solutions for addressing unmet clinical needs and improving patient outcome32,2.
5.3 Immunomodulatory proteins and their role in personalized medicine:
Immunomodulatory proteins have emerged as key players in the field of personalized medicine, facilitating the development of targeted immunotherapies for autoimmune diseases, inflammatory disorders, and cancer. Engineered proteins, such as checkpoint inhibitors, cytokine agonists, and chimeric antigen receptor (CAR) T-cell therapies, modulate the immune response and promote the selective targeting of diseased cells while preserving the integrity of healthy tissues.
The precise engineering of immunomodulatory proteins enables the customization of therapeutic interventions based on individual patient profiles, genetic predispositions, and disease progression patterns. These tailored approaches signify a significant advancement in the era of precision medicine, offering the potential for improved treatment outcomes and enhanced patient well-being 33.
5.4 Current challenges and future prospects in the clinical translation of engineered proteins:
Despite the remarkable progress in the development of engineered protein-based therapeutics, several challenges persist in their clinical translation and widespread adoption. Issues related to manufacturing scalability, regulatory approvals, and affordability pose hurdles to the seamless integration of these advanced applications into mainstream clinical practice.
However, the ongoing advancements in biotechnological processes, regulatory frameworks, and cost-effective manufacturing strategies offer promising solutions to overcome these challenges. The future prospects for engineered proteins in therapeutics remain bright, with continued research efforts and interdisciplinary collaborations poised to drive innovation and transform the landscape of modern healthcare8.
By exploring these advanced applications of engineered proteins in therapeutics, this section emphasizes the transformative potential of protein engineering in addressing complex medical challenges and fostering the development of personalized and precision medicine approaches.
This detailed section provides an extensive exploration of the advanced applications of engineered proteins in therapeutics, emphasizing their significance in targeted drug delivery, disease-specific treatments, and personalized immunomodulatory interventions. Discussion of current challenges and future prospects serves to underscore the opportunities and potential advancements in the clinical translation of engineered proteins for improved patient care and treatment outcomes33,10.
6. Regulatory considerations and safety profiles:
6.1 Regulatory guidelines for the development of engineered protein therapeutics:
The development and commercialization of engineered protein therapeutics necessitate adherence to stringent regulatory guidelines and standards to ensure patient safety, product efficacy, and manufacturing quality. Regulatory agencies, including the Food and Drug Administration (FDA), the European Medicines Agency (EMA), and other global regulatory bodies, outline specific requirements for preclinical testing, clinical trials, and post-marketing surveillance of engineered protein-based drugs.
These guidelines encompass rigorous assessments of product safety, pharmacokinetics, pharmacodynamics, and immunogenicity, as well as comprehensive evaluations of manufacturing processes and quality control measures. Compliance with these regulatory frameworks is essential for securing regulatory approvals and market authorizations, facilitating the safe and effective introduction of engineered protein therapeutics into clinical practice34.
6.2 Assessment of safety profiles and immunogenicity issues:
Ensuring the safety and tolerability of engineered protein therapeutics is a critical aspect of their clinical development and implementation. Evaluation of safety profiles encompasses comprehensive analyses of potential adverse reactions, toxicities, and off-target effects associated with the administration of engineered protein-based drugs in patient populations.
Immunogenicity issues represent a significant concern in the context of engineered protein therapeutics, as the introduction of foreign proteins may elicit immune responses, leading to the development of antibodies and immune-mediated adverse events. Rigorous preclinical and clinical assessments are conducted to characterize the immunogenic potential of engineered proteins, mitigate immune responses, and minimize the risk of treatment-associated complications35.
6.3 Preclinical and clinical evaluation of engineered protein-based drugs:
The preclinical and clinical evaluation of engineered protein-based drugs involves a systematic assessment of their pharmacological properties, safety profiles, and therapeutic efficacy in relevant disease models and patient cohorts. Preclinical studies focus on elucidating the pharmacokinetic and pharmacodynamic profiles, establishing dose-response relationships, and identifying potential safety concerns associated with the tested proteins.
Clinical trials, comprising multiple phases of rigorous testing in human subjects, aim to validate the safety, tolerability, and efficacy of engineered protein therapeutics. These trials assess parameters such as bioavailability, dosing regimens, and treatment outcomes, providing essential data for regulatory submissions and market approvals36.
6.4 Strategies for ensuring the efficacy and safety of engineered protein therapeutics:
The implementation of comprehensive strategies is paramount for ensuring the efficacy and safety of engineered protein therapeutics throughout their development, clinical testing, and post-marketing surveillance phases. These strategies encompass the integration of state-of-the-art analytical techniques, robust quality control measures, and standardized protocols for adverse event monitoring and reporting.
Moreover, the establishment of pharmacovigilance programs, post-marketing surveillance systems, and risk management plans facilitates the continuous assessment of the safety and efficacy profiles of marketed engineered protein-based drugs, ensuring timely detection and management of any potential safety concerns or product-related issues.
By emphasizing these regulatory considerations and safety profiles, this section underscores the critical importance of adhering to regulatory guidelines, ensuring comprehensive safety assessments, and implementing effective risk management strategies in the development and clinical translation of engineered protein therapeutics.
This detailed section offers a comprehensive exploration of regulatory considerations and safety profiles in the context of engineered protein therapeutics, emphasizing their significance in ensuring product safety, regulatory compliance, and patient well-being throughout the drug development lifecycle37.
7. Future directions and emerging trends in protein engineering:
7.1 Exploration of novel protein engineering techniques on the horizon:
The field of protein engineering continues to evolve, driven by ongoing research and technological advancements that hold the promise of unveiling novel techniques and methodologies. Emerging approaches, such as de novo protein design, computational protein redesign, and advanced gene editing technologies, offer exciting avenues for the development of next-generation engineered proteins with unprecedented functionalities and therapeutic applications.
These novel techniques aim to expand the boundaries of protein engineering, enabling the creation of bespoke proteins with tailored structures, functions, and properties to address previously unmet clinical needs and challenges. By fostering interdisciplinary collaborations and embracing cutting-edge technologies, researchers are poised to unlock the full potential of protein engineering in the pursuit of groundbreaking therapeutic solutions38,39.
7.2 Advancements in the application of artificial intelligence and machine learning:
The integration of artificial intelligence (AI) and machine learning (ML) has revolutionized the landscape of protein engineering, offering sophisticated tools for data analysis, predictive modeling, and protein design optimization. AI-driven algorithms facilitate the rapid processing of large-scale datasets, enabling researchers to discern intricate patterns, identify potential drug targets, and predict protein-ligand interactions with unparalleled accuracy.
Machine learning algorithms, trained on comprehensive protein databases and structural repositories, empower researchers to make informed decisions regarding protein modifications, structural predictions, and drug candidate selections. The application of AI and ML in protein engineering heralds a new era of precision medicine, personalized drug development, and tailored therapeutic interventions, paving the way for transformative advancements in the field40,41.
7.3 Potential implications for personalized and precision medicine:
The convergence of innovative protein engineering techniques, advanced computational modeling, and personalized medicine initiatives holds the potential to revolutionize the landscape of healthcare, ushering in an era of personalized and precision medicine tailored to individual patient profiles and genetic predispositions. Engineered proteins, customized to target specific disease pathways and molecular signatures, offer the promise of improved treatment outcomes, reduced side effects, and enhanced patient well-being.
The integration of personalized and precision medicine principles in the development of engineered protein therapeutics aims to address the heterogeneity of diseases, account for individual variations in treatment responses, and optimize therapeutic regimens based on patient-specific parameters. These tailored approaches signify a paradigm shift in the healthcare paradigm, emphasizing the importance of customized interventions and patient-centric care in the era of advanced protein engineering and biotechnology39,10.
7.4 Anticipated impact on the development of next-generation therapeutic interventions:
The anticipated impact of ongoing research and emerging trends in protein engineering is poised to revolutionize the development of next-generation therapeutic interventions, offering innovative solutions for a wide spectrum of diseases and medical conditions. From the design of novel protein-based biologics and cell therapies to the engineering of targeted drug delivery systems and regenerative medicine approaches, these advancements signify a transformative leap in the realm of modern healthcare.
The continual exploration of novel protein engineering techniques, the integration of AI-driven computational strategies, and the personalized and precision medicine initiatives collectively contribute to the development of safer, more effective, and patient-tailored therapeutic interventions. These advancements hold the potential to redefine treatment paradigms, improve clinical outcomes, and enhance the overall quality of life for patients worldwide.
By outlining these future directions and emerging trends, this section emphasizes the transformative potential of protein engineering in shaping the future of healthcare, underscoring the pivotal role of interdisciplinary research, technological innovation, and patient-centric approaches in driving the advancement of next-generation therapeutic interventions.
This detailed section provides a comprehensive exploration of future directions and emerging trends in protein engineering, emphasizing their significance in advancing personalized and precision medicine, harnessing the power of artificial intelligence, and fostering the development of transformative therapeutic interventions39,42.
8. CONCLUSION:
In conclusion, the field of protein engineering has emerged as a cornerstone in the development of advanced therapeutic interventions, offering transformative solutions for addressing complex medical challenges and improving patient outcomes. The comprehensive exploration of advanced techniques, including rational design, directed evolution, and computational modeling, has underscored the pivotal role of protein engineering in tailoring protein properties and functionalities to meet diverse clinical needs38,2.
Through the integration of rational design strategies, researchers have successfully enhanced protein stability, activity, and binding affinities, paving the way for the development of tailored protein-based therapeutics with improved efficacy and reduced off-target effects. Similarly, directed evolution techniques have facilitated the creation of diversified protein libraries and the identification of optimized variants with enhanced functionalities, demonstrating the versatility and transformative potential of this approach in therapeutic protein engineering.
The integration of computational modeling has expedited the protein engineering process, enabling the predictive analysis of protein structures, dynamics, and interactions, and facilitating the identification of potential drug candidates with high precision and efficiency. The synergistic interplay between computational and experimental approaches has demonstrated the transformative impact of computational modeling in guiding the rational design and optimization of therapeutic proteins for clinical applications.
The advanced applications of engineered proteins in targeted drug delivery, disease-specific treatments, and personalized immunomodulatory interventions signify a paradigm shift in the landscape of modern medicine, emphasizing the importance of precision and patient-centric approaches in the era of personalized and precision medicine.
While significant progress has been made in the field of protein engineering, several challenges persist in the clinical translation and widespread adoption of engineered protein-based therapeutics. Rigorous adherence to regulatory guidelines, comprehensive safety assessments, and effective risk management strategies are essential to ensure the safe and effective integration of these advanced applications into mainstream clinical practice9.
Looking ahead, the exploration of novel protein engineering techniques, the integration of AI-driven computational strategies, and the personalized and precision medicine initiatives hold the promise of revolutionizing the development of next-generation therapeutic interventions, fostering the advancement of tailored and patient-centric approaches in healthcare.
By highlighting these key findings and advancements in protein engineering, this comprehensive review aims to provide valuable insights for researchers, clinicians, and industry professionals, fostering the continued progress and application of advanced techniques in therapeutic biochemistry and biotechnology.
This detailed conclusion section provides a comprehensive recapitulation of the key findings and advancements in protein engineering, emphasizing their significance in personalized and precision medicine, the importance of regulatory compliance, and the transformative potential of future advancements in the field38,39.
9. CONFLICT OF INTEREST:
All others declare no relevant conflicts of interest. All authors contributed to the review and are responsible for the article content.
10. REFERENCES:
1. Faber MS, Whitehead TA. Data-driven engineering of protein therapeutics. Current Opinion in Biotechnology. 2019; Dec 1; 60: 104-10 https://doi.org/10.1016/j.copbio.2019.01.015
2. Singh DB, Tripathi T, editors. Protein-based Therapeutics. Springer Nature; 2023 Mar 1. https://doi.org/10.1007/978-981-19-8249-1
3. Carter PJ. Introduction to current and future protein therapeutics: a protein engineering perspective. Experimental Cell Research. 2011; May 15; 317(9): 1261-9. https://doi.org/10.1016/j.yexcr.2011.02.013
4. Li Y, Cirino PC. Recent advances in engineering proteins for biocatalysis. Biotechnology and Bioengineering. 2014; Jul; 111(7): 1273-87. https://doi.org/10.1002/bit.25240
5. Hellinga HW. Computational protein engineering. Nature Structural Biology. 1998; Jul; 5(7): 525-7. https://doi.org/10.1038/776
6. Qu G, Li A, Acevedo‐Rocha CG, Sun Z, Reetz MT. The crucial role of methodology development in directed evolution of selective enzymes. Angewandte Chemie International Edition. 2020; Aug 3; 59(32): 13204-31. https://doi.org/10.1002/anie.201901491
7. Kuhlman B, Bradley P. Advances in protein structure prediction and design. Nature Reviews Molecular Cell Biology. 2019; Nov; 20(11): 681-97. https://doi.org/10.1038/s41580-019-0163-x
8. Koga N, Tatsumi-Koga R, Liu G, Xiao R, Acton TB, Montelione GT, Baker D. Principles for designing ideal protein structures. Nature. 2012; Nov 8; 491(7423): 222-7. https://doi.org/10.1038/nature11600
9. S.D. Mankar, Bhagyashri S. Jadhav, Awantika N. Kale. Basics and Recent Advances in Peptide and Protein Drug Delivery. Asian Journal of Pharmacy and Technology. 2023; 13(3): 207-2 https://doi.org/10.52711/2231-5713.2023.00037
10. Akshay R. Yadav, Shrinivas K. Mohite. Recent Advances in Protein and Peptide Drug Delivery. Res. J. Pharma. Dosage Forms and Tech. 2020; 12(3): 205-212. https://doi.org/10.5958/0975-4377.2020.00035.X
11. Marshall SA, Lazar GA, Chirino AJ, Desjarlais JR. Rational design and engineering of therapeutic proteins. Drug Discovery Today. 2003; Mar 1; 8(5): 212-21. 9 https://doi.org/10.1016/S1359-6446(03)02610-2
12. Chevalier A, Silva DA, Rocklin GJ, Hicks DR, Vergara R, Murapa P, Bernard SM, Zhang L, Lam KH, Yao G, Bahl CD. Massively parallel de novo protein design for targeted therapeutics. Nature. 2017; Oct 5; 550(7674): 74-9. https://doi.org/10.1038/nature23912
13. Pace CN, Scholtz JM, Grimsley GR. Forces stabilizing proteins. FEBS Letters. 2014; Jun 27; 588(14): 2177-84. https://doi.org/10.1016/j.febslet.2014.05.006
14. Doyle SK, Pop MS, Evans HL, Koehler AN. Advances in discovering small molecules to probe protein function in a systems context. Current Opinion in Chemical Biology. 2016; Feb 1; 30: 28-36. https://doi.org/10.1016/j.cbpa.2015.10.032
15. Schreiber G, Fleishman SJ. Computational design of protein–protein interactions. Current opinion in structural biology. 2013; Dec 1; 23(6): 903-10. https://doi.org/10.1016/j.sbi.2013.08.003
16. Sameena Mehtab, Harpreet Parmar, Tanveer Irshad Siddiqi, Atanu Singha Roy. Determination of Protein Binding Affinities and Investigation into the Antimicrobial Activities of Cu(II), Co(II) and Ni(II) Mixed Ligand Complexes. Asian J. Research Chem. 2015; 8(2): February 99-107. https://doi.org/10.5958/0974-4150.2015.00018.8
17. Lane MD, Seelig B. Advances in the directed evolution of proteins. Current Opinion in Chemical Biology. 2014; Oct 1; 22: 129-36. https://doi.org/10.1016/j.cbpa.2014.09.013
18. Lutz S. Beyond directed evolution—semi-rational protein engineering and design. Current Opinion in Biotechnology. 2010; Dec 1; 21(6): 734-43.https://doi.org/10.1016/j.copbio.2010.08.011
19. Wójcik M, Telzerow A, Quax WJ, Boersma YL. High-throughput screening in protein engineering: recent advances and future perspectives. International Journal of Molecular Sciences. 2015; Oct 20; 16(10): 24918-45. https://doi.org/10.3390/ijms161024918
20. Vasserot AP, Dickinson CD, Tang Y, Huse WD, Manchester KS, Watkins JD. Optimization of protein therapeutics by directed evolution. Drug Discovery Today. 2003; Jan 21; 8(3): 118-26. https://doi.org/10.1016/S1359-6446(02)02590-4
21. Buchanan A, Ferraro F, Rust S, Sridharan S, Franks R, Dean G, McCourt M, Jermutus L, Minter R. Improved drug-like properties of therapeutic proteins by directed evolution. Protein Engineering, Design and Selection. 2012; Oct 1; 25(10): 631-8. https://doi.org/10.1093/protein/gzs054
22. Alka Singh, Bhaskar Kumar Gupta, Neeraj Upmanyu. A mini Review on Analytical and Bioanlytical Techniques in Study of Drugs-Protein Interaction. Asian Journal of Research in Chemistry. 2024; 17(2): 111-8. https://doi.org/10.52711/0974-4150.2024.00022
23. Susmi M S, Revathy S Kumar, Sreelakshmi V, Sruthy V Menon, Surya Mohan, Saranya Tulasidharan Suja, Sathianarayanan, Asha Asokan Manakadan. A Computational approach for identification of Phytochemicals for targeting and optimizing the inhibitors of Heat shock proteins. Research J. Pharm. and Tech. 2015; 8(9): Sept, 1199-1204 https://doi.org/10.5958/0974-360X.2015.00219.X
24. Derat E, Kamerlin SC. Computational advances in protein engineering and enzyme design. The Journal of Physical Chemistry B. 2022; Apr 7; 126(13): 2449-51. https://doi.org/10.1021/acs.jpcb.2c01198
25. Childers MC, Daggett V. Insights from molecular dynamics simulations for computational protein design. Molecular systems Design and Engineering. 2017; 2(1): 9-33. https://doi.org/10.1039/C6ME00083E
26. Akshay R. Yadav, Shrinivas K. Mohite. Homology Modeling and Generation of 3D-structure of Protein. Res. J. Pharma. Dosage Forms and Tech. 2020; 12(4): 313-320 https://doi.org/10.5958/0975-4377.2020.00052.X
27. Giordano D, Biancaniello C, Argenio MA, Facchiano A. Drug design by pharmacophore and virtual screening approach. Pharmaceuticals. 2022; May 23; 15(5): 646. https://doi.org/10.3390/ph15050646
28. Sathish Kumar Mittapalli, J N Narendra Sharath Chandra, Jay Prakash Soni, Ram Babu Tripathi, Iffath Rizwana. Virtual Screening and Pharmacophore Modeling for discovery of Biologically Active Natural Products as Inhibitors of Odorant-binding Proteins. Asian Journal of Research in Chemistry. 2024; 17(1): 17-4 https://doi.org/10.52711/0974-4150.2024.00004
29. Atkins JD, Boateng SY, Sorensen T, McGuffin LJ. Disorder prediction methods, their applicability to different protein targets and their usefulness for guiding experimental studies. International Journal of Molecular Sciences. 2015; Aug 13; 16(8): 19040-54. https://doi.org/10.3390/ijms160819040
30. Alvizo O, Allen BD, Mayo SL. Computational protein design promises to revolutionize protein engineering. Biotechniques. 2007; Jan; 42(1): 31-9. https://doi.org/10.2144/000112336
31. Elzoghby AO, Samy WM, Elgindy NA. Protein-based nanocarriers as promising drug and gene delivery systems. Journal of Controlled Release. 2012; Jul 10; 161(1): 38-49. https://doi.org/10.1016/j.jconrel.2012.04.036
32. MaHam A, Tang Z, Wu H, Wang J, Lin Y. Protein‐based nanomedicine platforms for drug delivery. Small. 2009; Aug 3; 5(15): 1706-21. https://doi.org/10.1002/smll.200801602
33. Wang G, Uludag H. Recent developments in nanoparticle-based drug delivery and targeting systems with emphasis on protein-based nanoparticles. Expert Opinion on Drug Delivery. 2008; May 1; 5(5): 499-515. https://doi.org/10.1517/17425247.5.5.499
34. Wang J, Shen WC, Zaro JL, editors. Antibody-drug conjugates: the 21st century magic bullets for cancer. New York, NY, USA: Springer; 2015; Mar 5. https://doi.org/10.1007/978-3-319-13081-1
35. Akash MS, Rehman K, Tariq M, Chen S. Development of therapeutic proteins: advances and challenges. Turkish Journal of Biology. 2015; 39(3): 343-58. https://doi.org/10.3906/biy-1411-8
36. Büttel IC, Chamberlain P, Chowers Y, Ehmann F, Greinacher A, Jefferis R, Kramer D, Kropshofer H, Lloyd P, Lubiniecki A, Krause R. Taking immunogenicity assessment of therapeutic proteins to the next level. Biologicals. 2011; Mar 1; 39(2): 100-9. https://doi.org/10.1016/j.biologicals.2011.01.006
37. Eon‐Duval A, Broly H, Gleixner R. Quality attributes of recombinant therapeutic proteins: an assessment of impact on safety and efficacy as part of a quality by design development approach. Biotechnology Progress. 2012; May; 28(3): 608-22. https://doi.org/10.1002/btpr.1548
38. Arindam Chakraborty, Dipak Kumar Singha, Manas Chakraborty, Payel Mukherjee. Protein Therapeutics: An Updated Review. Asian Journal of Research in Pharmaceutical Sciences. 2021; 11(3): 253-7. https://doi.org/10.52711/2231-5659.2021.00040
39. Namdev N., Upadhyay S. Challenges and approaches for Oral protein and peptide drug delivery. Research J. Pharm. and Tech. 2016; 9(3): Mar., 305-312. https://doi.org/10.5958/0974-360X.2016.00056.1
40. Leo DencelinX, Ramkumar T. Distributed Machine Learning Algorithms to classify Protein secondary structures for Drug Design – A Survey. Research J. Pharm. and Tech. 2017; 10(9): 3173-3180 https://doi.org/10.5958/0974-360X.2017.00564.9
41. Niazi SK, Mariam Z. Reinventing Therapeutic Proteins: Mining a treasure of new therapies. Biologics. 2023; Apr 19; 3(2): 72-94. https://doi.org/10.3390/biologics3020005
42. Caravella J, Lugovskoy A. Design of next-generation protein therapeutics. Current Opinion in Chemical Biology. 2010; Aug 1; 14(4): 520-8. https://doi.org/10.1016/j.cbpa.2010.06.175
Received on 30.07.2024 Revised on 20.11.2024 Accepted on 10.02.2025 Published on 01.07.2025 Available online from July 05, 2025 Research J. Pharmacy and Technology. 2025;18(7):3419-3427. DOI: 10.52711/0974-360X.2025.00493 © RJPT All right reserved
|
|
This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License. Creative Commons License. |
|