1
|
Zhou R, Qu J, Liu X, Lin F, Ohulchanskyy TY, Alifu N, Qu J, Yin DC. Biopharmaceutical drug delivery and phototherapy using protein crystals. Adv Drug Deliv Rev 2025; 216:115480. [PMID: 39613032 DOI: 10.1016/j.addr.2024.115480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Biopharmaceutical drugs, including proteins, peptides, and antibodies, are renowned for their high specificity and efficacy, fundamentally transforming disease treatment paradigms. However, their structural complexity presents challenges for their formulation and delivery. Protein crystals, characterized by high purity, high stability and a porous structure for biopharmaceutical drug encapsulation, providing a potential avenue for formulating and delivering biopharmaceutical drugs. There is increasing interest in engineering protein crystals to delivery biopharmaceutical drugs for biomedical applications. This review summarizes the recent advances in biopharmaceutical drug delivery and phototherapy using protein crystals. First, we evaluate the advantages of using protein crystals for biopharmaceutical drugs delivery. Next, we outline the strategies for in vitro and in vivo crystallization to prepare protein crystals. Importantly, the review highlights the advanced applications of protein crystals in biopharmaceutical drug delivery, tumor phototherapy, and other optical fields. Finally, it provides insights into future perspectives of biopharmaceutical drug delivery using protein crystals. This comprehensive review aims to provide effective insights into design of protein crystals to simplify biopharmaceutical drug delivery and improve disease treatment.
Collapse
Affiliation(s)
- Renbin Zhou
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Jinghan Qu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Xuejiao Liu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Fangrui Lin
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China.
| | - Tymish Y Ohulchanskyy
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Nuernisha Alifu
- School of Medical Engineering and Technology, Xinjiang Medical University, Urumqi 830054, China
| | - Junle Qu
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China; School of Medical Engineering and Technology, Xinjiang Medical University, Urumqi 830054, China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|
2
|
Singla D, Bhattacharya M. Preferential Binding of Cations Modulates Electrostatically Driven Protein Aggregation and Disaggregation. J Phys Chem B 2024; 128:10870-10879. [PMID: 39460751 DOI: 10.1021/acs.jpcb.4c06293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Protein aggregation resulting in either ordered amyloids or amorphous aggregates is not only restricted to deadly human diseases but also associated with biotechnological challenges encountered in the therapeutic and food industries. Elucidating the key structural determinants of protein aggregation is important to devise targeted inhibitory strategies, but it still remains a formidable task owing to the underlying hierarchy, stochasticity, and complexity associated with the self-assembly processes. Additionally, alterations in solution pH, salt types, and ionic strength modulate various noncovalent interactions, thus affecting the protein aggregation propensity and the aggregation kinetics. However, the molecular origin and a detailed understanding of the effects of weakly and strongly hydrated salts on protein aggregation and their plausible roles in the dissolution of aggregates remain elusive. In this study, using fluorescence and circular dichroism spectroscopy in combination with electron microscopy and light scattering techniques, we show that the ionic size, valency, and extent of hydration of cations play a crucial role in regulating the protein aggregation and disaggregation processes, which may elicit unique methods for governing the balance between protein self-assembly and disassembly.
Collapse
Affiliation(s)
- Deepika Singla
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India
| | - Mily Bhattacharya
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India
| |
Collapse
|
3
|
Jones W, Gerogiorgis DI. Dynamic optimization of an integrated cultivation-aggregation model for mAb production. Biotechnol Bioeng 2024; 121:2716-2727. [PMID: 38822680 DOI: 10.1002/bit.28761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
Due to their proteinaceous structure, monoclonal antibodies (mAbs) are susceptible to irreversible aggregation, with harmful consequences on drug efficacy and patient safety. To mitigate this risk in modern biopharmaceutical processes, it is critical to comply with current good manufacturing practices (cGMP) and pursue operating strategies minimizing irreversible aggregation whilst also maximizing mAb throughput. These conflicting objectives are targeted in this study by formulating and analyzing an integrated dynamic model accounting for both cultivation and aggregation of mAbs from a Chinese Hamster Ovary (CHO) cell line. Two manipulated dynamic variables are considered here in simulation studies: firstly temperature manipulation within a batch reactor, and secondly feed flow manipulation within a series of isothermal fed-batch reactors. Following this, dynamic optimization investigations have been conducted, firstly with the single objective of maximizing mAb throughput and secondly with multiple (two) objectives of maximizing mAb throughput while also minimizing irreversible aggregate content, simultaneously. The study provides key insight into tradeoffs of how simultaneous temperature and feed flowrate manipulation affects mAb throughput and aggregation inside bioreactors.
Collapse
Affiliation(s)
- Wil Jones
- School of Engineering, Institute for Materials and Processes (IMP), University of Edinburgh, Edinburgh, Scotland, UK
| | - Dimitrios I Gerogiorgis
- School of Engineering, Institute for Materials and Processes (IMP), University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
4
|
Panda C, Kumar S, Gupta S, Pandey LM. Insulin fibrillation under physicochemical parameters of bioprocessing and intervention by peptides and surface-active agents. Crit Rev Biotechnol 2024:1-22. [PMID: 39142855 DOI: 10.1080/07388551.2024.2387167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/23/2023] [Accepted: 06/17/2023] [Indexed: 08/16/2024]
Abstract
Even after the centenary celebration of insulin discovery, there prevail challenges concerning insulin aggregation, not only after repeated administration but also during industrial production, storage, transport, and delivery, significantly impacting protein quality, efficacy, and effectiveness. The aggregation reduces insulin bioavailability, increasing the risk of heightened immunogenicity, posing a threat to patient health, and creating a dent in the golden success story of insulin therapy. Insulin experiences various physicochemical and mechanical stresses due to modulations in pH, temperature, ionic strength, agitation, shear, and surface chemistry, during the upstream and downstream bioprocessing, resulting in insulin unfolding and subsequent fibrillation. This has fueled research in the pharmaceutical industry and academia to unveil the mechanistic insights of insulin aggregation in an attempt to devise rational strategies to regulate this unwanted phenomenon. The present review briefly describes the impacts of environmental factors of bioprocessing on the stability of insulin and correlates with various intermolecular interactions, particularly hydrophobic and electrostatic forces. The aggregation-prone regions of insulin are identified and interrelated with biophysical changes during stress conditions. The quest for novel additives, surface-active agents, and bioderived peptides in decelerating insulin aggregation, which results in overall structural stability, is described. We hope this review will help tackle the real-world challenges of insulin aggregation encountered during bioprocessing, ensuring safer, stable, and globally accessible insulin for efficient management of diabetes.
Collapse
Affiliation(s)
- Chinmaya Panda
- Bio-interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Sachin Kumar
- Viral Immunology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Sharad Gupta
- Neurodegeneration and Peptide Engineering Research Lab, Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, India
| | - Lalit M Pandey
- Bio-interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
5
|
Manning MC, Holcomb RE, Payne RW, Stillahn JM, Connolly BD, Katayama DS, Liu H, Matsuura JE, Murphy BM, Henry CS, Crommelin DJA. Stability of Protein Pharmaceuticals: Recent Advances. Pharm Res 2024; 41:1301-1367. [PMID: 38937372 DOI: 10.1007/s11095-024-03726-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024]
Abstract
There have been significant advances in the formulation and stabilization of proteins in the liquid state over the past years since our previous review. Our mechanistic understanding of protein-excipient interactions has increased, allowing one to develop formulations in a more rational fashion. The field has moved towards more complex and challenging formulations, such as high concentration formulations to allow for subcutaneous administration and co-formulation. While much of the published work has focused on mAbs, the principles appear to apply to any therapeutic protein, although mAbs clearly have some distinctive features. In this review, we first discuss chemical degradation reactions. This is followed by a section on physical instability issues. Then, more specific topics are addressed: instability induced by interactions with interfaces, predictive methods for physical stability and interplay between chemical and physical instability. The final parts are devoted to discussions how all the above impacts (co-)formulation strategies, in particular for high protein concentration solutions.'
Collapse
Affiliation(s)
- Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO, USA.
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA.
| | - Ryan E Holcomb
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Robert W Payne
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | | | | | | | | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | |
Collapse
|
6
|
Jin MJ, Ge XZ, Huang Q, Liu JW, Ingle RG, Gao D, Fang WJ. The Effects of Excipients on Freeze-dried Monoclonal Antibody Formulation Degradation and Sub-Visible Particle Formation during Shaking. Pharm Res 2024; 41:321-334. [PMID: 38291165 DOI: 10.1007/s11095-024-03657-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/09/2024] [Indexed: 02/01/2024]
Abstract
PURPOSES We previously reported an unexpected phenomenon that shaking stress could cause more protein degradation in freeze-dried monoclonal antibody (mAb) formulations than liquid ones (J Pharm Sci, 2022, 2134). The main purposes of the present study were to investigate the effects of shaking stress on protein degradation and sub-visible particle (SbVP) formation in freeze-dried mAb formulations, and to analyze the factors influencing protein degradation during production and transportation. METHODS The aggregation behavior of mAb-X formulations during production and transportation was simulated by shaking at a rate of 300 rpm at 25°C for 24 h. The contents of particles and monomers were analyzed by micro-flow imaging, dynamic light scattering, size exclusion chromatography, and ultraviolet - visible (UV-Vis) spectroscopy to compare the protective effects of excipients on the aggregation of mAb-X. RESULTS Shaking stress could cause protein degradation in freeze-dried mAb-X formulations, while surfactant, appropriate pH, polyol mannitol, and high protein concentration could impact SbVP generation. Water content had little effect on freeze-dried protein degradation during shaking, as far as the water content was controlled in the acceptable range as recommended by mainstream pharmacopoeias (i.e., less than 3%). CONCLUSIONS Shaking stress can reduce the physical stability of freeze-dried mAb formulations, and the addition of surfactants, polyol mannitol, and a high protein concentration have protective effects against the degradation of model mAb formulations induced by shaking stress. The experimental results provide new insight for the development of freeze-dried mAb formulations.
Collapse
Affiliation(s)
- Meng-Jia Jin
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Xin-Zhe Ge
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Qiong Huang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Jia-Wei Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Rahul G Ingle
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education & Research, Sawangi, Wardha, India
| | - Dong Gao
- Zhejiang Bioray Biopharmaceutical Co., Taizhou, 317000, China
| | - Wei-Jie Fang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China.
- Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua, 321000, China.
- Taizhou Institute of Zhejiang University, Taizhou, 317000, China.
| |
Collapse
|
7
|
Panda C, Sharma LG, Pandey LM. Experimental procedures to investigate fibrillation of proteins. MethodsX 2023; 11:102445. [PMID: 37928109 PMCID: PMC10622682 DOI: 10.1016/j.mex.2023.102445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023] Open
Abstract
The unwanted phenomenon of protein fibrillation is observed in vivo and during therapeutic protein development in the industry. Protein aggregation is associated with various degenerative disorders and might induce immune-related challenges post-administration of biopharmaceutics. A pipeline for early detection, identification, and removal of pre-formed fibrils is needed to improve the quality, efficacy, and effectiveness of the formulation. Protein fibril formation is accompanied by unfolding, secondary structural changes and the formation of larger aggregates. However, most detection processes come with extensive sample preparation steps and inefficient repeatability, incurring a financial burden on research. The current article summarizes and critically discusses six simple yet powerful methods to detect aggregation phenomena in the line of detecting fibrillar aggregates in heat-induced bovine serum albumin protein. Comparing the native and heat-induced protein samples would provide insights about aggregates. Easy, inexpensive and optimized protocols for detecting the fibrillation of proteins are explained. The procedures mentioned here detected the appearance of β-sheet-rich fibrils in the heat-induced protein sample. The aggregation is characterized by enhanced thioflavin-T fluorescence, alteration in the intrinsic fluorescence, decrease in helicity and subsequent increase in β-sheet and appearance of particles with larger hydrodynamic diameters. •This article summarizes various analytical techniques to easily characterize the fibrillation of proteins.•Various techniques to detect the formation of β-sheet rich structures, changes in the secondary structures and size of aggregates have been discussed.•The stated methodologies are validated on a model protein, albumin.
Collapse
Affiliation(s)
- Chinmaya Panda
- Bio-interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Laipubam Gayatri Sharma
- Bio-interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Lalit M Pandey
- Bio-interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| |
Collapse
|
8
|
Vitharana S, Stillahn JM, Katayama DS, Henry CS, Manning MC. Application of Formulation Principles to Stability Issues Encountered During Processing, Manufacturing, and Storage of Drug Substance and Drug Product Protein Therapeutics. J Pharm Sci 2023; 112:2724-2751. [PMID: 37572779 DOI: 10.1016/j.xphs.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
The field of formulation and stabilization of protein therapeutics has become rather extensive. However, most of the focus has been on stabilization of the final drug product. Yet, proteins experience stress and degradation through the manufacturing process, starting with fermentaition. This review describes how formulation principles can be applied to stabilize biopharmaceutical proteins during bioprocessing and manufacturing, considering each unit operation involved in prepration of the drug substance. In addition, the impact of the container on stabilty is discussed as well.
Collapse
Affiliation(s)
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
9
|
Elsayed A, Jaber N, Al-Remawi M, Abu-Salah K. From cell factories to patients: Stability challenges in biopharmaceuticals manufacturing and administration with mitigation strategies. Int J Pharm 2023; 645:123360. [PMID: 37657507 DOI: 10.1016/j.ijpharm.2023.123360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/19/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023]
Abstract
Active ingredients of biopharmaceuticals consist of a wide array of biomolecular structures, including those of enzymes, monoclonal antibodies, nucleic acids, and recombinant proteins. Recently, these molecules have dominated the pharmaceutical industry owing to their safety and efficacy. However, their manufacturing is hindered by high cost, inadequate batch-to-batch equivalence, inherent instability, and other quality issues. This article is an up-to-date review of the challenges encountered during different stages of biopharmaceutical production and mitigation of problems arising during their development, formulation, manufacturing, and administration. It is a broad overview discussion of stability issues encountered during product life cycle i.e., upstream processing (aggregation, solubility, host cell proteins, color change), downstream bioprocessing (aggregation, fragmentation), formulation, manufacturing, and delivery to patients.
Collapse
Affiliation(s)
- Amani Elsayed
- College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Nisrein Jaber
- Faculty of Pharmacy, Al Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Mayyas Al-Remawi
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman 1196, Jordan.
| | - Khalid Abu-Salah
- King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Department of Nanomedicine, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Panda C, Kumar S, Gupta S, Pandey LM. Structural, kinetic, and thermodynamic aspects of insulin aggregation. Phys Chem Chem Phys 2023; 25:24195-24213. [PMID: 37674360 DOI: 10.1039/d3cp03103a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Given the significance of protein aggregation in proteinopathies and the development of therapeutic protein pharmaceuticals, revamped interest in assessing and modelling the aggregation kinetics has been observed. Quantitative analysis of aggregation includes data of gradual monomeric depletion followed by the formation of subvisible particles. Kinetic and thermodynamic studies are essential to gain key insights into the aggregation process. Despite being the medical marvel in the world of diabetes, insulin suffers from the challenge of aggregation. Physicochemical stresses are experienced by insulin during industrial formulation, storage, delivery, and transport, considerably impacting product quality, efficacy, and effectiveness. The present review briefly describes the pathways, mathematical kinetic models, and thermodynamics of protein misfolding and aggregation. With a specific focus on insulin, further discussions include the structural heterogeneity and modifications of the intermediates incurred during insulin fibrillation. Finally, different model equations to fit the kinetic data of insulin fibrillation are discussed. We believe that this review will shed light on the conditions that induce structural changes in insulin during the lag phase of fibrillation and will motivate scientists to devise strategies to block the initialization of the aggregation cascade. Subsequent abrogation of insulin fibrillation during bioprocessing will ensure stable and globally accessible insulin for efficient management of diabetes.
Collapse
Affiliation(s)
- Chinmaya Panda
- Bio-interface & Environmental Engineering Lab Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| | - Sachin Kumar
- Viral Immunology Lab Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India
| | - Sharad Gupta
- Neurodegeneration and Peptide Engineering Research Lab Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Gujarat, 382355, India
| | - Lalit M Pandey
- Bio-interface & Environmental Engineering Lab Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| |
Collapse
|
11
|
Sonbhadra S, Mehak, Pandey LM. Biogenesis, Isolation, and Detection of Exosomes and Their Potential in Therapeutics and Diagnostics. BIOSENSORS 2023; 13:802. [PMID: 37622888 PMCID: PMC10452587 DOI: 10.3390/bios13080802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/26/2023]
Abstract
The increasing research and rapid developments in the field of exosomes provide insights into their role and significance in human health. Exosomes derived from various sources, such as mesenchymal stem cells, cardiac cells, and tumor cells, to name a few, can be potential therapeutic agents for the treatment of diseases and could also serve as biomarkers for the early detection of diseases. Cellular components of exosomes, several proteins, lipids, and miRNAs hold promise as novel biomarkers for the detection of various diseases. The structure of exosomes enables them as drug delivery vehicles. Since exosomes exhibit potential therapeutic applications, their efficient isolation from complex biological/clinical samples and precise real-time analysis becomes significant. With the advent of microfluidics, nano-biosensors are being designed to capture exosomes efficiently and rapidly. Herein, we have summarized the history, biogenesis, characteristics, functions, and applications of exosomes, along with the isolation, detection, and quantification techniques. The implications of surface modifications to enhance specificity have been outlined. The review also sheds light on the engineered nanoplatforms being developed for exosome detection and capture.
Collapse
Affiliation(s)
| | | | - Lalit M. Pandey
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (S.S.); (M.)
| |
Collapse
|
12
|
Sant D, Rojekar S, Gera S, Pallapati A, Gimenez-Roig J, Kuo TC, Padilla A, Korkmaz F, Cullen L, Chatterjee J, Shelly E, Meseck M, Miyashita S, Macdonald A, Sultana F, Barak O, Ryu V, Kim SM, Robinson C, Rosen CJ, Caminis J, Lizneva D, Haider S, Yuen T, Zaidi M. Optimizing a therapeutic humanized follicle-stimulating hormone-blocking antibody formulation by protein thermal shift assay. Ann N Y Acad Sci 2023; 1521:67-78. [PMID: 36628526 PMCID: PMC11658029 DOI: 10.1111/nyas.14952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Biopharmaceutical products are formulated using several Food and Drug Administration (FDA) approved excipients within the inactive ingredient limit to maintain their storage stability and shelf life. Here, we have screened and optimized different sets of excipient combinations to yield a thermally stable formulation for the humanized follicle-stimulating hormone (FSH)-blocking antibody, MS-Hu6. We used a protein thermal shift assay in which rising temperatures resulted in the maximal unfolding of the protein at the melting temperature (Tm ). To determine the buffer and pH for a stable solution, four different buffers with a pH range from 3 to 8 were screened. This resulted in maximal Tm s at pH 5.62 for Fab in phosphate buffer and at pH 6.85 for Fc in histidine buffer. Upon testing a range of salt concentrations, MS-Hu6 was found to be more stable at lower concentrations, likely due to reduced hydrophobic effects. Molecular dynamics simulations revealed a higher root-mean-square deviation with 1 mM than with 100 mM salt, indicating enhanced stability, as noted experimentally. Among the stabilizers tested, Tween 20 was found to yield the highest Tm and reversed the salt effect. Among several polyols/sugars, trehalose and sucrose were found to produce higher thermal stabilities. Finally, binding of recombinant human FSH to MS-Hu6 in a final formulation (20 mM phosphate buffer, 1 mM NaCl, 0.001% w/v Tween 20, and 260 mM trehalose) resulted in a thermal shift (increase in Tm ) for the Fab, but expectedly not in the Fc domain. Given that we used a low dose of MS-Hu6 (1 μM), the next challenge would be to determine whether 100-fold higher, industry-standard concentrations are equally stable.
Collapse
Affiliation(s)
- Damini Sant
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Satish Rojekar
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Sakshi Gera
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Anusha Pallapati
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Judit Gimenez-Roig
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Tan-Chun Kuo
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Ashley Padilla
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Liam Cullen
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Jiya Chatterjee
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Eleanor Shelly
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Marcia Meseck
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Anne Macdonald
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Farhath Sultana
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Orly Barak
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Se-Min Kim
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Cemre Robinson
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Clifford J. Rosen
- Maine Medical Center Research Institute, Scarborough, Maine 040774, USA
| | - John Caminis
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Shozeb Haider
- UCL Centre for Advanced Research Computing, School of Pharmacy, London, WC1N 1AX, UK
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| |
Collapse
|
13
|
Mahanta N, Sharma S, Sharma LG, Pandey LM, Dixit US. Unfolding of the SARS-CoV-2 spike protein through infrared and ultraviolet-C radiation based disinfection. Int J Biol Macromol 2022; 221:71-82. [PMID: 36063893 PMCID: PMC9439869 DOI: 10.1016/j.ijbiomac.2022.08.197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/12/2022] [Accepted: 08/30/2022] [Indexed: 11/05/2022]
Abstract
The spreading of coronavirus from contacting surfaces and aerosols created a pandemic around the world. To prevent the transmission of SARS-CoV-2 virus and other contagious microbes, disinfection of contacting surfaces is necessary. In this study, a disinfection box equipped with infrared (IR) radiation heating and ultraviolet-C (UV-C) radiation is designed and tested for its disinfection ability against pathogenic bacteria and SARS-CoV-2 spike protein. The killing of a Gram-positive, namely, S. aureus and a Gram-negative namely, S. typhi bacteria was studied followed by the inactivation of the spike protein. The experimental parameters were optimized using a statistical tool. For the broad-spectrum antibacterial activity, the optimum condition was holding at 65.61 °C for 13.54 min. The killing of the bacterial pathogen occurred via rupturing the cell walls as depicted by electron microscopy. Further, the unfolding of SARS-CoV-2 spike protein and RNase A was studied under IR and UV-C irradiations at the aforesaid optimized condition. The unfolding of both the proteins was confirmed by changes in the secondary structure, particularly an increase in β-sheets and a decrease in α-helixes. Remarkably, the higher penetration depth of IR waves up to subcutaneous tissue resulted in lower optimum disinfection temperature, <70 °C in vogue. Thus, the combined UV-C and IR radiation is effective in killing the pathogenic bacteria and denaturing the glycoproteins.
Collapse
Affiliation(s)
- Nilkamal Mahanta
- Department of Mechanical Engineering, Indian Institute of Technology Guwahati, India
| | - Swati Sharma
- Bio-Interface and Environmental Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, India
| | - Laipubam Gayatri Sharma
- Bio-Interface and Environmental Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, India
| | - Lalit M Pandey
- Bio-Interface and Environmental Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, India
| | - Uday Shanker Dixit
- Department of Mechanical Engineering, Indian Institute of Technology Guwahati, India.
| |
Collapse
|