1
|
Rahban M, Ahmad F, Piatyszek MA, Haertlé T, Saso L, Saboury AA. Stabilization challenges and aggregation in protein-based therapeutics in the pharmaceutical industry. RSC Adv 2023; 13:35947-35963. [PMID: 38090079 PMCID: PMC10711991 DOI: 10.1039/d3ra06476j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/30/2023] [Indexed: 04/26/2024] Open
Abstract
Protein-based therapeutics have revolutionized the pharmaceutical industry and become vital components in the development of future therapeutics. They offer several advantages over traditional small molecule drugs, including high affinity, potency and specificity, while demonstrating low toxicity and minimal adverse effects. However, the development and manufacturing processes of protein-based therapeutics presents challenges related to protein folding, purification, stability and immunogenicity that should be addressed. These proteins, like other biological molecules, are prone to chemical and physical instabilities. The stability of protein-based drugs throughout the entire manufacturing, storage and delivery process is essential. The occurrence of structural instability resulting from misfolding, unfolding, and modifications, as well as aggregation, poses a significant risk to the efficacy of these drugs, overshadowing their promising attributes. Gaining insight into structural alterations caused by aggregation and their impact on immunogenicity is vital for the advancement and refinement of protein therapeutics. Hence, in this review, we have discussed some features of protein aggregation during production, formulation and storage as well as stabilization strategies in protein engineering and computational methods to prevent aggregation.
Collapse
Affiliation(s)
- Mahdie Rahban
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences Kerman Iran
| | - Faizan Ahmad
- Department of Biochemistry, School of Chemical & Life Sciences, Jamia Hamdard New Delhi-110062 India
| | | | | | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University Rome Italy
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran Tehran 1417614335 Iran +9821 66404680 +9821 66956984
| |
Collapse
|
2
|
Lafi Z, Gharaibeh L, Nsairat H, Asha N, Alshaer W. Aptasensors: employing molecular probes for precise medical diagnostics and drug monitoring. Bioanalysis 2023; 15:1439-1460. [PMID: 37847048 DOI: 10.4155/bio-2023-0141] [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] [Indexed: 10/18/2023] Open
Abstract
Accurate detection and monitoring of therapeutic drug levels are vital for effective patient care and treatment management. Aptamers, composed of single-stranded DNA or RNA molecules, are integral components of biosensors designed for both qualitative and quantitative detection of biological samples. Aptasensors play crucial roles in target identification, validation, detection of drug-target interactions and screening potential of drug candidates. This review focuses on the pivotal role of aptasensors in early disease detection, particularly in identifying biomarkers associated with various diseases such as cancer, infectious diseases and cardiovascular disorders. Aptasensors have demonstrated exceptional potential in enhancing disease diagnostics and monitoring therapeutic drug levels. Aptamer-based biosensors represent a transformative technology in the field of healthcare, enabling precise diagnostics, drug monitoring and disease detection.
Collapse
Affiliation(s)
- Zainab Lafi
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Lobna Gharaibeh
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Hamdi Nsairat
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Nisreen Asha
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
3
|
Batugal T, Pendyala G, Tomasovic L, Varner C, Caplin JD, Page AM, Davis M, Satola SW, García AJ, Kane RS. Engineering active lysostaphin variants that incorporate noncanonical amino acids and characterizing the effects of site-specific PEGylation. Biotechnol Bioeng 2023; 120:1694-1701. [PMID: 36810983 DOI: 10.1002/bit.28360] [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: 12/07/2022] [Revised: 02/11/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023]
Abstract
We describe a facile strategy to identify sites for the incorporation of noncanonical amino acids into lysostaphin-an enzyme that degrades the cell wall of Staphylococcus aureus-while retaining stapholytic activity. We used this strategy to generate active variants of lysostaphin incorporating para-azidophenylalanine. The incorporation of this "reactive handle" enabled the orthogonal site-specific modification of the enzyme variants with polyethylene glycol (PEG) using copper-free click cycloaddition. PEGylated lysostaphin variants could retain their stapholytic activity, with the extent of retention depending on the site of modification and the PEG molecular weight. The site-specific modification of lysostaphin could be useful not only for PEGylation to improve biocompatibility but also for the incorporation of the enzyme into hydrogels and other biomaterials and for studies of protein structure and dynamics. Moreover, the approach described herein could be readily applied to identify suitable sites for the incorporation of reactive handles into other proteins of interest.
Collapse
Affiliation(s)
- Troy Batugal
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Geetanjali Pendyala
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Luke Tomasovic
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Chad Varner
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Jeremy D Caplin
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Alexander M Page
- Department of Medicine, Division of Infectious Diseases and Emory Investigational Clinical Microbiology Core, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michelle Davis
- Department of Medicine, Division of Infectious Diseases and Emory Investigational Clinical Microbiology Core, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sarah W Satola
- Department of Medicine, Division of Infectious Diseases and Emory Investigational Clinical Microbiology Core, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Ravi S Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Schijven LM, Saggiomo V, Velders AH, Bitter JH, Nikiforidis CV. On the influence of protein aggregate sizes for the formation of solid and hollow protein microparticles. J Colloid Interface Sci 2022; 631:181-190. [DOI: 10.1016/j.jcis.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
|
5
|
Yeo XY, Cunliffe G, Ho RC, Lee SS, Jung S. Potentials of Neuropeptides as Therapeutic Agents for Neurological Diseases. Biomedicines 2022; 10:343. [PMID: 35203552 PMCID: PMC8961788 DOI: 10.3390/biomedicines10020343] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Despite recent leaps in modern medicine, progress in the treatment of neurological diseases remains slow. The near impermeable blood-brain barrier (BBB) that prevents the entry of therapeutics into the brain, and the complexity of neurological processes, limits the specificity of potential therapeutics. Moreover, a lack of etiological understanding and the irreversible nature of neurological conditions have resulted in low tolerability and high failure rates towards existing small molecule-based treatments. Neuropeptides, which are small proteinaceous molecules produced by the body, either in the nervous system or the peripheral organs, modulate neurological function. Although peptide-based therapeutics originated from the treatment of metabolic diseases in the 1920s, the adoption and development of peptide drugs for neurological conditions are relatively recent. In this review, we examine the natural roles of neuropeptides in the modulation of neurological function and the development of neurological disorders. Furthermore, we highlight the potential of these proteinaceous molecules in filling gaps in current therapeutics.
Collapse
Affiliation(s)
- Xin Yi Yeo
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore; (X.Y.Y.); (G.C.)
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Grace Cunliffe
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore; (X.Y.Y.); (G.C.)
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Roger C. Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Institute for Health Innovation & Technology (iHealthtech), National University of Singapore, Singapore 117599, Singapore
| | - Su Seong Lee
- NanoBio Lab, Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore
| | - Sangyong Jung
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore; (X.Y.Y.); (G.C.)
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|
6
|
Giri P, Pagar AD, Patil MD, Yun H. Chemical modification of enzymes to improve biocatalytic performance. Biotechnol Adv 2021; 53:107868. [PMID: 34774927 DOI: 10.1016/j.biotechadv.2021.107868] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/23/2022]
Abstract
Improvement in intrinsic enzymatic features is in many instances a prerequisite for the scalable applicability of many industrially important biocatalysts. To this end, various strategies of chemical modification of enzymes are maturing and now considered as a distinct way to improve biocatalytic properties. Traditional chemical modification methods utilize reactivities of amine, carboxylic, thiol and other side chains originating from canonical amino acids. On the other hand, noncanonical amino acid- mediated 'click' (bioorthogoal) chemistry and dehydroalanine (Dha)-mediated modifications have emerged as an alternate and promising ways to modify enzymes for functional enhancement. This review discusses the applications of various chemical modification tools that have been directed towards the improvement of functional properties and/or stability of diverse array of biocatalysts.
Collapse
Affiliation(s)
- Pritam Giri
- Department of Systems Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Amol D Pagar
- Department of Systems Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Mahesh D Patil
- Department of Nanomaterials and Application Technology, Center of Innovative and Applied Bioprocessing (CIAB), Sector-81, PO Manauli, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Hyungdon Yun
- Department of Systems Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
7
|
Vaghi L, Monti M, Marelli M, Motto E, Papagni A, Cipolla L. Photoinduced Porcine Gelatin Cross-Linking by Homobi- and Homotrifunctional Tetrazoles. Gels 2021; 7:124. [PMID: 34449602 PMCID: PMC8395868 DOI: 10.3390/gels7030124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
Gelatin is a costless polypeptide material of natural origin, able to form hydrogels that are potentially useful in biomaterial scaffold design for drug delivery, cell cultures, and tissue engineering. However, gelatin hydrogels are unstable at physiological conditions, losing their features only after a few minutes at 37 °C. Accordingly, treatments to address this issue are of great interest. In the present work, we propose for the first time the use of bi- and trifunctional tetrazoles, most of them unknown to date, for photoinduced gelatin cross-linking towards the production of physiologically stable hydrogels. Indeed, after UV-B irradiation, aryl tetrazoles generate a nitrilimine intermediate that is reactive towards different functionalities, some of them constitutively present in the amino acid side chains of gelatin. The efficacy of the treatment strictly depends on the structure of the cross-linking agent used, and substantial improved stability was observed by switching from bifunctional to trifunctional cross-linkers.
Collapse
Affiliation(s)
- Luca Vaghi
- Dipartimento di Scienza dei Materiali, Università degli Studi di Milano—Bicocca, via R. Cozzi 55, 20125 Milano, Italy;
| | - Mauro Monti
- Dipartimento di Scienza dei Materiali, Università degli Studi di Milano—Bicocca, via R. Cozzi 55, 20125 Milano, Italy;
| | - Marcello Marelli
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, CNR-SCITEC, Sede Fantoli, via Fantoli 16/15, 20138 Milano, Italy;
| | - Elisa Motto
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano—Bicocca, Piazza della Scienza 2, 20126 Milano, Italy;
| | - Antonio Papagni
- Dipartimento di Scienza dei Materiali, Università degli Studi di Milano—Bicocca, via R. Cozzi 55, 20125 Milano, Italy;
| | - Laura Cipolla
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano—Bicocca, Piazza della Scienza 2, 20126 Milano, Italy;
| |
Collapse
|
8
|
Synthesis, LC-MS/MS analysis, and biological evaluation of two vaccine candidates against ticks based on the antigenic P0 peptide from R. sanguineus linked to the p64K carrier protein from Neisseria meningitidis. Anal Bioanal Chem 2021; 413:5885-5900. [PMID: 34341841 PMCID: PMC8328535 DOI: 10.1007/s00216-021-03569-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 11/24/2022]
Abstract
A peptide from the P0 acidic ribosomal protein (pP0) of ticks conjugated to keyhole limpet hemocyanin from Megathura crenulata has shown to be effective against different tick species when used in host vaccination. Turning this peptide into a commercial anti-tick vaccine will depend on finding the appropriate, technically and economically feasible way to present it to the host immune system. Two conjugates (p64K-Cys1pP0 and p64K-βAla1pP0) were synthesized using the p64K carrier protein from Neisseria meningitidis produced in Escherichia coli, the same cross-linking reagent, and two analogues of pP0. The SDS-PAGE analysis of p64K-Cys1pP0 showed a heterogeneous conjugate compared to p64K-βAla1pP0 that was detected as a protein band at 91kDa. The pP0/p64K ratio determined by MALDI-MS for p64K-Cys1pP0 ranged from 1 to 8, being 3-5 the predominant ratio, while in the case of p64K-βAla1pP0 this ratio was 5-7. Cys1pP0 was partially linked to 35 out of 39 Lys residues and the N-terminal end, while βAla1pP0 was mostly linked to the six free cysteine residues, to the N-terminal end, and, in a lesser extent, to Lys residues. The assignment of the conjugation sites and side reactions were based on the identification of type 2 peptides. Rabbit immunizations showed the best anti-pP0 titers and the highest efficacy against Rhipicephalus sanguineus ticks when the p64K-Cys1pP0 was used as vaccine antigen. The presence of high molecular mass aggregates observed in the SDS-PAGE analysis of p64K-Cys1pP0 could be responsible for a better immune response against pP0 and consequently for its better efficacy as an anti-tick vaccine. Graphical abstract ![]()
Collapse
|
9
|
New insights into affinity proteins for HER2-targeted therapy: Beyond trastuzumab. Biochim Biophys Acta Rev Cancer 2020; 1874:188448. [PMID: 33039514 DOI: 10.1016/j.bbcan.2020.188448] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 12/31/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is known as a potential target for both cancer treatment and diagnosis. One of the most interesting HER2-targeted therapeutics is an affinity protein which selectively recognizes and binds to a defined target. Trastuzumab is a monoclonal antibody which has been approved as the first affinity proteins for treatment of some HER2-positive cancers including breast cancer. Despite initial response to trastuzumab, the majority of patients with metastatic HER2-positive breast cancer still show resistance to the therapy. Recently, various anti-HER2 affinity proteins, including antibodies, antibody fragments (e.g., Fab and scFv) and other protein scaffolds (e.g., affibody and DARPin), alone or fused/conjugated with therapeutic agents (e.g., proteins, drugs and radioisotopes) have been developed to overcome the trastuzumab resistance. Here, we review these engineered affinity proteins which are either clinically approved or under evaluation. Modern technologies and future prospects for their clinical applications in cancer treatment are also discussed.
Collapse
|
10
|
Sarkar S, Gulati K, Mishra A, Poluri KM. Protein nanocomposites: Special inferences to lysozyme based nanomaterials. Int J Biol Macromol 2020; 151:467-482. [DOI: 10.1016/j.ijbiomac.2020.02.179] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/15/2020] [Accepted: 02/16/2020] [Indexed: 12/19/2022]
|
11
|
Ling X, Xie B, Gao X, Chang L, Zheng W, Chen H, Huang Y, Tan L, Li M, Liu T. Improving the efficiency of precise genome editing with site-specific Cas9-oligonucleotide conjugates. SCIENCE ADVANCES 2020; 6:eaaz0051. [PMID: 32494588 PMCID: PMC7250679 DOI: 10.1126/sciadv.aaz0051] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/13/2020] [Indexed: 05/02/2023]
Abstract
Site-specific chemical conjugation of proteins can enhance their therapeutic and diagnostic utility but has seldom been applied to CRISPR-Cas9, which is a rapidly growing field with great therapeutic potential. The low efficiency of homology-directed repair remains a major hurdle in CRISPR-Cas9-mediated precise genome editing, which is limited by low concentration of donor DNA template at the cleavage site. In this study, we have developed methodology to site-specifically conjugate oligonucleotides to recombinant Cas9 protein containing a genetically encoded noncanonical amino acid with orthogonal chemical reactivity. The Cas9-oligonucleotide conjugates recruited an unmodified donor DNA template to the target site through base pairing, markedly increasing homology-directed repair efficiency in both human cell culture and mouse zygotes. These chemically modified Cas9 mutants provide an additional tool, one that is complementary to chemically modified nucleic acids, for improving the utility of CRISPR-Cas9-based genome-editing systems.
Collapse
Affiliation(s)
- Xinyu Ling
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Bingteng Xie
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, 100191 Beijing, China
| | - Xiaoqin Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Liying Chang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Wei Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Heqi Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yujia Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Linzhi Tan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Mo Li
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, 100191 Beijing, China
- Corresponding author. (T.L.); (M.L.)
| | - Tao Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- Corresponding author. (T.L.); (M.L.)
| |
Collapse
|
12
|
Doti N, Caporale A, Monti A, Sandomenico A, Selis F, Ruvo M. A recent update on the use of microbial transglutaminase for the generation of biotherapeutics. World J Microbiol Biotechnol 2020; 36:53. [PMID: 32172335 DOI: 10.1007/s11274-020-02829-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/07/2020] [Indexed: 01/12/2023]
Abstract
The recent scientific progresses on the use of enzyme-mediated reactions in organic, non-aqueous and aqueous media have significantly supported the growing demand of new biotechnological and/or pharmacological products. Today, a plethora of microbial enzymes, used as biocatalysts, are available. Among these, microbial transglutaminases (MTGs) are broadly used for their ability to catalyse the formation of an isopeptide bond between the γ-amide group of glutamines and the ε-amino group of lysine. Due to their promiscuity towards primary amine-containing substrates and the more stringent specificity for glutamine-containing peptide sequences, several combined approaches can be tailored for different settings, making MTGs very attractive catalysts for generating protein-protein and protein small molecule's conjugates. The present review offers a recent update on the modifications attainable by MTG-catalysed bioreactions as reported between 2014 and 2019. In particular, we present a detailed and comparative overview on the MTG-based methods for proteins and antibodies engineering, with a particular outlook on the synthesis of homogeneous antibody-drug conjugates.
Collapse
Affiliation(s)
- N Doti
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.
| | - A Caporale
- Institute of Crystallography, CNR (IC-CNR), c/o Area Science Park s.s. 14 Km 163.5, Basovizza, 34149, Trieste, Italy
| | - Alessandra Monti
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABIF), University L. Vanvitelli, Via Vivaldi, 43, 80100, Caserta, Italy
| | - A Sandomenico
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy
| | - Fabio Selis
- BioVIIIx R&D, Via B. Brin, 59C, 80142, Naples, Italy
| | - M Ruvo
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.
| |
Collapse
|
13
|
Berckman EA, Hartzell EJ, Mitkas AA, Sun Q, Chen W. Biological Assembly of Modular Protein Building Blocks as Sensing, Delivery, and Therapeutic Agents. Annu Rev Chem Biomol Eng 2020; 11:35-62. [PMID: 32155350 DOI: 10.1146/annurev-chembioeng-101519-121526] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nature has evolved a wide range of strategies to create self-assembled protein nanostructures with structurally defined architectures that serve a myriad of highly specialized biological functions. With the advent of biological tools for site-specific protein modifications and de novo protein design, a wide range of customized protein nanocarriers have been created using both natural and synthetic biological building blocks to mimic these native designs for targeted biomedical applications. In this review, different design frameworks and synthetic decoration strategies for achieving these functional protein nanostructures are summarized. Key attributes of these designer protein nanostructures, their unique functions, and their impact on biosensing and therapeutic applications are discussed.
Collapse
Affiliation(s)
- Emily A Berckman
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA; .,Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Emily J Hartzell
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA;
| | - Alexander A Mitkas
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA;
| | - Qing Sun
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA;
| |
Collapse
|
14
|
Better together: building protein oligomers naturally and by design. Biochem Soc Trans 2020; 47:1773-1780. [PMID: 31803901 PMCID: PMC6925524 DOI: 10.1042/bst20190283] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 12/17/2022]
Abstract
Protein oligomers are more common in nature than monomers, with dimers being the most prevalent final structural state observed in known structures. From a biological perspective, this makes sense as it conserves vital molecular resources that may be wasted simply by generating larger single polypeptide units, and allows new features such as cooperativity to emerge. Taking inspiration from nature, protein designers and engineers are now building artificial oligomeric complexes using a variety of approaches to generate new and useful supramolecular protein structures. Oligomerisation is thus offering a new approach to sample structure and function space not accessible through simply tinkering with monomeric proteins.
Collapse
|
15
|
Singh G, Zarschler K, Hunoldt S, Martínez IIS, Ruehl CL, Matterna M, Bergmann R, Máthé D, Hegedüs N, Bachmann M, Comba P, Stephan H. Versatile Bispidine-Based Bifunctional Chelators for 64 Cu II -Labelling of Biomolecules. Chemistry 2020; 26:1989-2001. [PMID: 31755596 PMCID: PMC7028042 DOI: 10.1002/chem.201904654] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Indexed: 12/11/2022]
Abstract
Bifunctional chelators as parts of modular metal-based radiopharmaceuticals are responsible for stable complexation of the radiometal ion and for covalent linkage between the complex and the targeting vector. To avoid loss of complex stability, the bioconjugation strategy should not interfere with the radiometal chelation by occupying coordinating groups. The C9 position of the very stable CuII chelator 3,7-diazabicyclo[3.3.1]nonane (bispidine) is virtually predestined to introduce functional groups for facile bioconjugation as this functionalisation does not disturb the metal binding centre. We describe the preparation and characterisation of a set of novel bispidine derivatives equipped with suitable functional groups for diverse bioconjugation reactions, including common amine coupling strategies (bispidine-isothiocyanate) and the Cu-free strain-promoted alkyne-azide cycloaddition. We demonstrate their functionality and versatility in an exemplary way by conjugation to an antibody-based biomolecule and validate the obtained conjugate in vitro and in vivo.
Collapse
Affiliation(s)
- Garima Singh
- Helmholtz-Zentrum Dresden-RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Kristof Zarschler
- Helmholtz-Zentrum Dresden-RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Sebastian Hunoldt
- Helmholtz-Zentrum Dresden-RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Irma Ivette Santana Martínez
- Helmholtz-Zentrum Dresden-RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Carmen L. Ruehl
- Anorganisch-Chemisches Institut INF 270Universität Heidelberg69120HeidelbergGermany
| | - Madlen Matterna
- Helmholtz-Zentrum Dresden-RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Domokos Máthé
- Department of Biophysics and Radiation BiologySemmelweis University1094BudapestHungary
- CROmed Translational Research Centers Ltd.1047BudapestHungary
| | - Nikolett Hegedüs
- Department of Biophysics and Radiation BiologySemmelweis University1094BudapestHungary
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| | - Peter Comba
- Anorganisch-Chemisches Institut INF 270Universität Heidelberg69120HeidelbergGermany
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-RossendorfInstitute of Radiopharmaceutical Cancer ResearchBautzner Landstrasse 40001328DresdenGermany
| |
Collapse
|
16
|
Zhou F, Fu T, Huang Q, Kuai H, Mo L, Liu H, Wang Q, Peng Y, Han D, Zhao Z, Fang X, Tan W. Hypoxia-Activated PEGylated Conditional Aptamer/Antibody for Cancer Imaging with Improved Specificity. J Am Chem Soc 2019; 141:18421-18427. [PMID: 31584808 DOI: 10.1021/jacs.9b05063] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Aptamers and antibodies, as molecular recognition probes, play critical roles in cancer diagnosis and therapy. However, their recognition ability is based on target overexpression in disease cells, not target exclusivity, which can cause on-target off-tumor effects. To address the limitation, we herein report a novel strategy to develop a conditional aptamer conjugate which recognizes its cell surface target, but only after selective activation, as determined by characteristics of the disease microenvironment, which, in our model, involve tumor hypoxia. This conditional aptamer is the result of conjugating the aptamer with PEG5000-azobenzene-NHS, which is responsive to hypoxia, here acting as a caging moiety of conditional recognition. More specifically, the caging moiety is unresponsive in the intact conjugate and prevents target recognition. However, in the presence of sodium dithionite or hypoxia (<0.1% O2) or in the tumor microenvironment, the caging moiety responds by allowing conditional recognition of the cell-surface target, thereby reducing the chance of on-target off-tumor effects. It is also confirmed that the strategy can be used for developing a conditional antibody. Therefore, this study demonstrates an efficient strategy by which to develop aptamer/antibody-based diagnostic probes and therapeutic drugs for cancers with a unique hypoxic microenvironment.
Collapse
Affiliation(s)
- Fang Zhou
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Ting Fu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Qin Huang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Hailan Kuai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Liuting Mo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Honglin Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Qianqian Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Yongbo Peng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Dongmei Han
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Zilong Zhao
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China
| | - Xiaohong Fang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , P. R. China.,Institute of Cancer and Basic Medicine , Chinese Academy of Sciences (IBMC); Cancer Hospital, University of Chinese Academy of Sciences , Hangzhou , Zhejiang 310022 , China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , P. R. China.,Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering , Shanghai Jiao Tong University Shanghai , Shanghai , P. R. China.,Institute of Cancer and Basic Medicine , Chinese Academy of Sciences (IBMC); Cancer Hospital, University of Chinese Academy of Sciences , Hangzhou , Zhejiang 310022 , China
| |
Collapse
|
17
|
Friedman Ohana R, Hurst R, Rosenblatt M, Levin S, Machleidt T, Kirkland TA, Encell LP, Robers MB, Wood KV. Utilizing a Simple Method for Stoichiometric Protein Labeling to Quantify Antibody Blockade. Sci Rep 2019; 9:7046. [PMID: 31065015 PMCID: PMC6504924 DOI: 10.1038/s41598-019-43469-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Ligand binding assays routinely employ fluorescently-labeled protein ligands to quantify the extent of binding. These ligands are commonly generated through chemical modification of accessible lysine residues, which often results in heterogeneous populations exhibiting variable binding properties. This could be remedied by quantitative, site-specific labeling. Recently, we reported on a single-step method integrating recombinant protein purification with 2-cyanobenzothiazole (CBT) condensation for labeling a proteolytically exposed N-terminal cysteine. Here, using three growth factors, we show that unlike random lysine labeling, this site-specific approach yielded homogeneous populations of growth factors that were quantitatively labeled at their N-termini and retained their binding characteristics. We demonstrate the utility of this labeling method through the development of a novel assay that quantifies the capacity of antibodies to block receptor-ligand interactions (i.e. antibody blockade). The assay uses bioluminescence resonance energy transfer (BRET) to detect binding of CBT-labeled growth factors to their cognate receptors genetically fused to NanoLuc luciferase. The ability of antibodies to block these interactions is quantified through decrease in BRET. Using several antibodies, we show that the assay provides reliable quantification of antibody blockade in a cellular context. As demonstrated here, this simple method for generating uniformly-labeled proteins has potential to promote more accurate and robust ligand binding assays.
Collapse
Affiliation(s)
| | - Robin Hurst
- Promega Corporation, 2800 Woods Hollow Rd, Madison, WI, 53711, USA
| | - Mike Rosenblatt
- Promega Corporation, 2800 Woods Hollow Rd, Madison, WI, 53711, USA
| | - Sergiy Levin
- Promega Biosciences LLC, 277 Granada Dr, San Luis Obispo, CA, 93401, USA
| | - Thomas Machleidt
- Promega Corporation, 2800 Woods Hollow Rd, Madison, WI, 53711, USA
| | - Thomas A Kirkland
- Promega Biosciences LLC, 277 Granada Dr, San Luis Obispo, CA, 93401, USA
| | - Lance P Encell
- Promega Corporation, 2800 Woods Hollow Rd, Madison, WI, 53711, USA
| | - Matthew B Robers
- Promega Corporation, 2800 Woods Hollow Rd, Madison, WI, 53711, USA
| | - Keith V Wood
- Promega Corporation, 2800 Woods Hollow Rd, Madison, WI, 53711, USA
| |
Collapse
|
18
|
Yong KW, Yuen D, Chen MZ, Porter CJH, Johnston APR. Pointing in the Right Direction: Controlling the Orientation of Proteins on Nanoparticles Improves Targeting Efficiency. NANO LETTERS 2019; 19:1827-1831. [PMID: 30773887 DOI: 10.1021/acs.nanolett.8b04916] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Protein-conjugated nanoparticles have the potential to precisely deliver therapeutics to target sites in the body by specifically binding to cell surface receptors. To maximize targeting efficiency, the three-dimensional presentation of ligands toward these receptors is crucial. Herein, we demonstrate significantly enhanced targeting of nanoparticles to cancer cells by controlling the protein orientation on the nanoparticle surface. To engineer the point of attachment, we used amber codon reassignment to incorporate a synthetic amino acid, p-azidophenylalanine (azPhe), at specific locations within a single domain antibody (sdAb or nanobody) that recognizes the human epidermal growth factor receptor (EGFR). The azPhe modified sdAb can be tethered to the nanoparticle in a specific orientation using a bioorthogonal click reaction with a strained cyclooctyne. The crystal structure of the sdAb bound to EGFR was used to rationally select sites likely to optimally display the sdAb upon conjugation to a fluorescent nanocrystal (Qdot). Qdots with sdAb attached at the azPhe13 position showed 6 times greater binding affinity to EGFR expressing A549 cells, compared to Qdots with conventionally (succinimidyl ester) conjugated sdAb. As ligand-targeted delivery systems move toward clinical application, this work shows that nanoparticle targeting can be optimized by engineering the site of protein conjugation.
Collapse
Affiliation(s)
- Ken W Yong
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria 3052 , Australia
| | - Daniel Yuen
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria 3052 , Australia
| | - Moore Z Chen
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria 3052 , Australia
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria 3052 , Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , Monash University , Parkville , Victoria 3052 , Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria 3052 , Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , Monash University , Parkville , Victoria 3052 , Australia
| |
Collapse
|
19
|
Wang Y, Dong M, Ye M. A chemoenzymatic approach enables the site-specific conjugation of recombinant proteins. Electrophoresis 2019; 40:2125-2128. [PMID: 30793337 DOI: 10.1002/elps.201800528] [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: 12/26/2018] [Revised: 02/09/2019] [Accepted: 02/10/2019] [Indexed: 11/05/2022]
Abstract
Many biotechniques including protein microarray, drug screening, biosensors rely on the immobilization of recombinant proteins on the solid supports. It is well known that random orientation of the immobilized proteins could impair their biologic functions. Thus, it is very important to develop new site-specific immobilization approach. In this study, we presented a chemoenzymatic approach for site-specific conjugation of recombinant proteins onto solid support. In this strategy, the affinity tag on recombinant protein was enzymatically cleaved to expose the N-terminal serine, which was oxidized to carry an aldehyde group and was then covalently coupled to hydrazide resin through hydrazone ligation. As this approach takes advantage of the most frequently used TEV protease, it requires no further sequence design on recombinant protein. This method was validated by site specific coupling of a synthetic peptide and a recombinant protein onto solid supports. It was found that the site specific immobilized SH2 domain is functional and could be used to enrich tyrosine phosphorylated peptides.
Collapse
Affiliation(s)
- Yan Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, University of Chinese Academy of Sciences, Beijing, P. R. China.,National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, P. R. China
| | - Mingming Dong
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, University of Chinese Academy of Sciences, Beijing, P. R. China.,Dalian Ocean University, Dalian, P. R. China
| | - Mingliang Ye
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, University of Chinese Academy of Sciences, Beijing, P. R. China.,National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, P. R. China
| |
Collapse
|
20
|
Akgun B, Hall DG. Boronic Acids as Bioorthogonal Probes for Site‐Selective Labeling of Proteins. Angew Chem Int Ed Engl 2018; 57:13028-13044. [DOI: 10.1002/anie.201712611] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/23/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Burcin Akgun
- Department of Chemistry—CCIS 4–010University of Alberta Edmonton Alberta T6G 2G2 Canada
| | - Dennis G. Hall
- Department of Chemistry—CCIS 4–010University of Alberta Edmonton Alberta T6G 2G2 Canada
| |
Collapse
|
21
|
Akgun B, Hall DG. Boronsäuren als bioorthogonale Sonden für zentrenselektives Protein‐Labeling. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201712611] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Burcin Akgun
- Department of Chemistry – CCIS 4-010University of Alberta Edmonton Alberta T6G 2G2 Kanada
| | - Dennis G. Hall
- Department of Chemistry – CCIS 4-010University of Alberta Edmonton Alberta T6G 2G2 Kanada
| |
Collapse
|
22
|
Yates NDJ, Fascione MA, Parkin A. Methodologies for "Wiring" Redox Proteins/Enzymes to Electrode Surfaces. Chemistry 2018; 24:12164-12182. [PMID: 29637638 PMCID: PMC6120495 DOI: 10.1002/chem.201800750] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Indexed: 12/22/2022]
Abstract
The immobilization of redox proteins or enzymes onto conductive surfaces has application in the analysis of biological processes, the fabrication of biosensors, and in the development of green technologies and biochemical synthetic approaches. This review evaluates the methods through which redox proteins can be attached to electrode surfaces in a "wired" configuration, that is, one that facilitates direct electron transfer. The feasibility of simple electroactive adsorption onto a range of electrode surfaces is illustrated, with a highlight on the recent advances that have been achieved in biotechnological device construction using carbon materials and metal oxides. The covalent crosslinking strategies commonly used for the modification and biofunctionalization of electrode surfaces are also evaluated. Recent innovations in harnessing chemical biology methods for electrically wiring redox biology to surfaces are emphasized.
Collapse
Affiliation(s)
| | | | - Alison Parkin
- Department of ChemistryUniversity of YorkHeslington RoadYorkYO10 5DDUK
| |
Collapse
|
23
|
Lee TH, Hirst DJ, Kulkarni K, Del Borgo MP, Aguilar MI. Exploring Molecular-Biomembrane Interactions with Surface Plasmon Resonance and Dual Polarization Interferometry Technology: Expanding the Spotlight onto Biomembrane Structure. Chem Rev 2018; 118:5392-5487. [PMID: 29793341 DOI: 10.1021/acs.chemrev.7b00729] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The molecular analysis of biomolecular-membrane interactions is central to understanding most cellular systems but has emerged as a complex technical challenge given the complexities of membrane structure and composition across all living cells. We present a review of the application of surface plasmon resonance and dual polarization interferometry-based biosensors to the study of biomembrane-based systems using both planar mono- or bilayers or liposomes. We first describe the optical principals and instrumentation of surface plasmon resonance, including both linear and extraordinary transmission modes and dual polarization interferometry. We then describe the wide range of model membrane systems that have been developed for deposition on the chips surfaces that include planar, polymer cushioned, tethered bilayers, and liposomes. This is followed by a description of the different chemical immobilization or physisorption techniques. The application of this broad range of engineered membrane surfaces to biomolecular-membrane interactions is then overviewed and how the information obtained using these techniques enhance our molecular understanding of membrane-mediated peptide and protein function. We first discuss experiments where SPR alone has been used to characterize membrane binding and describe how these studies yielded novel insight into the molecular events associated with membrane interactions and how they provided a significant impetus to more recent studies that focus on coincident membrane structure changes during binding of peptides and proteins. We then discuss the emerging limitations of not monitoring the effects on membrane structure and how SPR data can be combined with DPI to provide significant new information on how a membrane responds to the binding of peptides and proteins.
Collapse
Affiliation(s)
- Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Daniel J Hirst
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Mark P Del Borgo
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| |
Collapse
|
24
|
Khoshnejad M, Greineder CF, Pulsipher KW, Villa CH, Altun B, Pan DC, Tsourkas A, Dmochowski IJ, Muzykantov VR. Ferritin Nanocages with Biologically Orthogonal Conjugation for Vascular Targeting and Imaging. Bioconjug Chem 2018; 29:1209-1218. [PMID: 29429330 DOI: 10.1021/acs.bioconjchem.8b00004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Genetic incorporation of biologically orthogonal functional groups into macromolecules has the potential to yield efficient, controlled, reproducible, site-specific conjugation of affinity ligands, contrast agents, or therapeutic cargoes. Here, we applied this approach to ferritin, a ubiquitous iron-storage protein that self-assembles into multimeric nanocages with remarkable stability, size uniformity (12 nm), and endogenous capacity for loading and transport of a variety of inorganic and organic cargoes. The unnatural amino acid, 4-azidophenylalanine (4-AzF), was incorporated at different sites in the human ferritin light chain (hFTL) to allow site-specific conjugation of alkyne-containing small molecules or affinity ligands to the exterior surface of the nanocage. The optimal positioning of the 4-AzF residue was evaluated by screening a library of variants for the efficiency of copper-free click conjugation. One of the engineered ferritins, hFTL-5X, was found to accommodate ∼14 small-molecule fluorophores (AlexaFluor 488) and 3-4 IgG molecules per nanocage. Intravascular injection in mice of radiolabeled hFTL-5X carrying antibody to cell adhesion molecule ICAM-1, but not control IgG, enabled specific targeting to the lung due to high basal expression of ICAM-1 (43.3 ± 6.99 vs 3.48 ± 0.14%ID/g for Ab vs IgG). Treatment of mice with endotoxin known to stimulate inflammatory ICAM-1 overexpression resulted in 2-fold enhancement of pulmonary targeting (84.4 ± 12.89 vs 43.3 ± 6.99%ID/g). Likewise, injection of fluorescent, ICAM-targeted hFTL-5X nanocages revealed the effect of endotoxin by enhancement of near-infrared signal, indicating potential utility of this approach for both vascular targeting and imaging.
Collapse
|
25
|
Affiliation(s)
- Madduri Srinivasarao
- Purdue Institute for Drug
Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Philip S. Low
- Purdue Institute for Drug
Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
26
|
Marth G, Hartley AM, Reddington SC, Sargisson LL, Parcollet M, Dunn KE, Jones DD, Stulz E. Precision Templated Bottom-Up Multiprotein Nanoassembly through Defined Click Chemistry Linkage to DNA. ACS NANO 2017; 11:5003-5010. [PMID: 28414900 DOI: 10.1021/acsnano.7b01711] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We demonstrate an approach that allows attachment of single-stranded DNA (ssDNA) to a defined residue in a protein of interest (POI) so as to provide optimal and well-defined multicomponent assemblies. Using an expanded genetic code system, azido-phenylalanine (azF) was incorporated at defined residue positions in each POI; copper-free click chemistry was used to attach exactly one ssDNA at precisely defined residues. By choosing an appropriate residue, ssDNA conjugation had minimal impact on protein function, even when attached close to active sites. The protein-ssDNA conjugates were used to (i) assemble double-stranded DNA systems with optimal communication (energy transfer) between normally separate groups and (ii) generate multicomponent systems on DNA origami tiles, including those with enhanced enzyme activity when bound to the tile. Our approach allows any potential protein to be simply engineered to attach ssDNA or related biomolecules, creating conjugates for designed and highly precise multiprotein nanoscale assembly with tailored functionality.
Collapse
Affiliation(s)
- Gabriella Marth
- School of Chemistry and Institute for Life Sciences, University of Southampton , Highfield, Southampton SO17 1BJ, United Kingdom
| | - Andrew M Hartley
- School of Biosciences, Cardiff University , Cardiff CF10 3AT, United Kingdom
| | - Samuel C Reddington
- School of Biosciences, Cardiff University , Cardiff CF10 3AT, United Kingdom
| | - Lauren L Sargisson
- School of Chemistry and Institute for Life Sciences, University of Southampton , Highfield, Southampton SO17 1BJ, United Kingdom
| | - Marlène Parcollet
- School of Chemistry and Institute for Life Sciences, University of Southampton , Highfield, Southampton SO17 1BJ, United Kingdom
| | - Katherine E Dunn
- Department of Electronics, University of York , Heslington, York YO10 5DD, United Kingdom
| | - D Dafydd Jones
- School of Biosciences, Cardiff University , Cardiff CF10 3AT, United Kingdom
| | - Eugen Stulz
- School of Chemistry and Institute for Life Sciences, University of Southampton , Highfield, Southampton SO17 1BJ, United Kingdom
| |
Collapse
|
27
|
Meyer JP, Adumeau P, Lewis JS, Zeglis BM. Click Chemistry and Radiochemistry: The First 10 Years. Bioconjug Chem 2016; 27:2791-2807. [PMID: 27787983 DOI: 10.1021/acs.bioconjchem.6b00561] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The advent of click chemistry has had a profound influence on almost all branches of chemical science. This is particularly true of radiochemistry and the synthesis of agents for positron emission tomography (PET), single photon emission computed tomography (SPECT), and targeted radiotherapy. The selectivity, ease, rapidity, and modularity of click ligations make them nearly ideally suited for the construction of radiotracers, a process that often involves working with biomolecules in aqueous conditions with inexorably decaying radioisotopes. In the following pages, our goal is to provide a broad overview of the first 10 years of research at the intersection of click chemistry and radiochemistry. The discussion will focus on four areas that we believe underscore the critical advantages provided by click chemistry: (i) the use of prosthetic groups for radiolabeling reactions, (ii) the creation of coordination scaffolds for radiometals, (iii) the site-specific radiolabeling of proteins and peptides, and (iv) the development of strategies for in vivo pretargeting. Particular emphasis will be placed on the four most prevalent click reactions-the Cu-catalyzed azide-alkyne cycloaddition (CuAAC), the strain-promoted azide-alkyne cycloaddition (SPAAC), the inverse electron demand Diels-Alder reaction (IEDDA), and the Staudinger ligation-although less well-known click ligations will be discussed as well. Ultimately, it is our hope that this review will not only serve to educate readers but will also act as a springboard, inspiring synthetic chemists and radiochemists alike to harness click chemistry in even more innovative and ambitious ways as we embark upon the second decade of this fruitful collaboration.
Collapse
Affiliation(s)
| | - Pierre Adumeau
- Department of Chemistry, Hunter College of the City University of New York , 413 East 69th Street, New York, New York 10028, United States
| | - Jason S Lewis
- Department of Radiology, Weill Cornell Medical College , 520 East 70th Street, New York, New York 10065, United States
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York , 413 East 69th Street, New York, New York 10028, United States.,Department of Radiology, Weill Cornell Medical College , 520 East 70th Street, New York, New York 10065, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , 365 5th Avenue, New York, New York 10016, United States
| |
Collapse
|
28
|
Ta DT, Guedens W, Vranken T, Vanschoenbeek K, Steen Redeker E, Michiels L, Adriaensens P. Enhanced Biosensor Platforms for Detecting the Atherosclerotic Biomarker VCAM1 Based on Bioconjugation with Uniformly Oriented VCAM1-Targeting Nanobodies. BIOSENSORS-BASEL 2016; 6:bios6030034. [PMID: 27399790 PMCID: PMC5039653 DOI: 10.3390/bios6030034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 01/05/2023]
Abstract
Surface bioconjugation of biomolecules has gained enormous attention for developing advanced biomaterials including biosensors. While conventional immobilization (by physisorption or covalent couplings using the functional groups of the endogenous amino acids) usually results in surfaces with low activity, reproducibility and reusability, the application of methods that allow for a covalent and uniformly oriented coupling can circumvent these limitations. In this study, the nanobody targeting Vascular Cell Adhesion Molecule-1 (NbVCAM1), an atherosclerotic biomarker, is engineered with a C-terminal alkyne function via Expressed Protein Ligation (EPL). Conjugation of this nanobody to azidified silicon wafers and Biacore™ C1 sensor chips is achieved via Copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) “click” chemistry to detect VCAM1 binding via ellipsometry and surface plasmon resonance (SPR), respectively. The resulting surfaces, covered with uniformly oriented nanobodies, clearly show an increased antigen binding affinity, sensitivity, detection limit, quantitation limit and reusability as compared to surfaces prepared by random conjugation. These findings demonstrate the added value of a combined EPL and CuAAC approach as it results in strong control over the surface orientation of the nanobodies and an improved detecting power of their targets—a must for the development of advanced miniaturized, multi-biomarker biosensor platforms.
Collapse
Affiliation(s)
- Duy Tien Ta
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Diepenbeek BE-3590, Belgium.
- Faculty of Food Technology and Biotechnology, Can Tho University of Technology, Can Tho 900000, Vietnam.
| | - Wanda Guedens
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Diepenbeek BE-3590, Belgium.
| | - Tom Vranken
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Diepenbeek BE-3590, Belgium.
| | - Katrijn Vanschoenbeek
- Immunology and Biochemistry, Biomedical Research Institute (Biomed) and School of Life Sciences, Transnationale Universiteit Limburg, Hasselt University, Diepenbeek BE-3590, Belgium.
| | - Erik Steen Redeker
- Maastricht Science Programme, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Luc Michiels
- Immunology and Biochemistry, Biomedical Research Institute (Biomed) and School of Life Sciences, Transnationale Universiteit Limburg, Hasselt University, Diepenbeek BE-3590, Belgium.
| | - Peter Adriaensens
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Diepenbeek BE-3590, Belgium.
- Applied and Analytical Chemistry, Institute for Materials Research (IMO), Hasselt University, Diepenbeek BE-3590, Belgium.
| |
Collapse
|
29
|
Maruani A, Richards DA, Chudasama V. Dual modification of biomolecules. Org Biomol Chem 2016; 14:6165-78. [PMID: 27278999 DOI: 10.1039/c6ob01010e] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
With the advent of novel bioorthogonal reactions and "click" chemistry, an increasing number of strategies for the single labelling of proteins and oligonucleotides have emerged. Whilst several methods exist for the site-selective introduction of a single chemical moiety, site-selective and bioorthogonal dual modification of biomolecules remains a challenge. The introduction of multiple modules enables a plethora of permutations and combinations and can generate a variety of bioconjuguates with many potential applications. From de novo approaches on oligomers to the post-translational functionalisation of proteins, this review will highlight the main strategies to dually modify biomolecules.
Collapse
Affiliation(s)
- Antoine Maruani
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H OAJ, UK.
| | | | | |
Collapse
|
30
|
Schatte M, Bocola M, Roth T, Martinez R, Kopetzki E, Schwaneberg U, Bönitz-Dulat M. Reporter Immobilization Assay (REIA) for Bioconjugating Reactions. Bioconjug Chem 2016; 27:1484-92. [DOI: 10.1021/acs.bioconjchem.6b00111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Martin Schatte
- Lehrstuhl
für Biotechnologie, RWTH Aachen University, 52074 Aachen, Germany
| | - Marco Bocola
- Lehrstuhl
für Biotechnologie, RWTH Aachen University, 52074 Aachen, Germany
| | | | - Ronny Martinez
- Lehrstuhl
für Biotechnologie, RWTH Aachen University, 52074 Aachen, Germany
| | | | - Ulrich Schwaneberg
- Lehrstuhl
für Biotechnologie, RWTH Aachen University, 52074 Aachen, Germany
| | | |
Collapse
|
31
|
Si L, Li P, Liu X, Luo L. Chinese herb medicine against Sortase A catalyzed transformations, a key role in gram-positive bacterial infection progress. J Enzyme Inhib Med Chem 2016; 31:184-196. [PMID: 27162091 DOI: 10.1080/14756366.2016.1178639] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Many Gram-positive bacteria can anchor their surface proteins to the cell wall peptidoglycan covalently by a common mechanism with Sortase A (SrtA), thus escaping from the host's identification of immune cells. SrtA can complete this anchoring process by cleaving LPXTG motif conserved among these surface proteins and thus these proteins anchor on the cell wall. Moreover, those SrtA mutants lose this capability to anchor these relative proteins, with these bacteria no longer infectious. Therefore, SrtA inhibitors can be promising anti-infective agents to cure bacterial infections. Chinese herb medicines (CHMs) (chosen from Science Citation Index) have exhibited inhibition on SrtA of Gram-positive pathogens irreversibly or reversibly. In general, CHMs are likely to have important long-term impact as new antibacterial compounds and sought after by academia and the pharmaceutical industry. This review mainly focuses on SrtA inhibitors from CHMs and the potential inhibiting mechanism related to chemical structures of compounds in CHMs.
Collapse
Affiliation(s)
- Lifang Si
- a School of Bioscience & Bioengineering, South China University of Technology, Guangzhou University Town , Panyu , Guangzhou , China
| | - Pan Li
- a School of Bioscience & Bioengineering, South China University of Technology, Guangzhou University Town , Panyu , Guangzhou , China
| | - Xiong Liu
- a School of Bioscience & Bioengineering, South China University of Technology, Guangzhou University Town , Panyu , Guangzhou , China
| | - Lixin Luo
- a School of Bioscience & Bioengineering, South China University of Technology, Guangzhou University Town , Panyu , Guangzhou , China
| |
Collapse
|
32
|
Ta DT, Redeker ES, Billen B, Reekmans G, Sikulu J, Noben JP, Guedens W, Adriaensens P. An efficient protocol towards site-specifically clickable nanobodies in high yield: cytoplasmic expression in Escherichia coli combined with intein-mediated protein ligation. Protein Eng Des Sel 2015; 28:351-63. [PMID: 26243885 DOI: 10.1093/protein/gzv032] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/01/2015] [Indexed: 11/13/2022] Open
Abstract
In this study, several expression strategies were investigated in order to develop a generic, highly productive and efficient protocol to produce nanobodies modified with a clickable alkyne function at their C-terminus via the intein-mediated protein ligation (IPL) technique. Hereto, the nanobody targeting the vascular cell adhesion molecule 1 (NbVCAM1) was used as a workhorse. The highlights of the protocol can be ascribed to a cytoplasmic expression of the nanobody-intein-chitin-binding domain fusion protein in the Escherichia coli SHuffle(®) T7 cells with a C-terminal extension, i.e. LEY, EFLEY or His6 spacer peptide, in the commonly used Luria-Bertani medium. The combination of these factors led to a high yield (up to 22 mg/l of culture) and nearly complete alkynation efficiency of the C-terminally modified nanobody via IPL. This yield can even be improved to ∼45 mg/l in the EnPresso(®) growth system but this method is more expensive and time-consuming. The resulting alkynated nanobodies retained excellent binding capacity towards the recombinant human VCAM1. The presented protocol benefits from time- and cost-effectiveness, which allows a feasible production up-scaling of generic alkynated nanobodies. The production of high quantities of site-specifically modified nanobodies paves the way to new biosurface applications that demand for a homogeneously oriented nanobody coupling. Prospectively, the alkynated nanobodies can be covalently coupled to a multitude of azide-containing counterparts, e.g. contrast labeling agents, particles or surfaces for numerous innovative applications.
Collapse
Affiliation(s)
- Duy Tien Ta
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium Faculty of Food Technology and Biotechnology, Can Tho University of Technology, Can Tho, Vietnam
| | - Erik Steen Redeker
- Maastricht Science Programme, Maastricht University, Maastricht 6200 MD, The Netherlands
| | - Brecht Billen
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| | - Gunter Reekmans
- Applied and Analytical Chemistry, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| | - Josephine Sikulu
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| | - Jean-Paul Noben
- Biomedical Research Institute (Biomed) and School of Life Sciences, Transnationale Universiteit Limburg, Hasselt University, Agoralaan-Building C, Diepenbeek BE-3590, Belgium
| | - Wanda Guedens
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| | - Peter Adriaensens
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium Applied and Analytical Chemistry, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| |
Collapse
|
33
|
Eckmann DM, Composto RJ, Tsourkas A, Muzykantov VR. Nanogel Carrier Design for Targeted Drug Delivery. J Mater Chem B 2014; 2:8085-8097. [PMID: 25485112 PMCID: PMC4251498 DOI: 10.1039/c4tb01141d] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Polymer-based nanogel formulations offer features attractive for drug delivery, including ease of synthesis, controllable swelling and viscoelasticity as well as drug loading and release characteristics, passive and active targeting, and the ability to formulate nanogel carriers that can respond to biological stimuli. These unique features and low toxicity make the nanogels a favorable option for vascular drug targeting. In this review, we address key chemical and biological aspects of nanogel drug carrier design. In particular, we highlight published studies of nanogel design, descriptions of nanogel functional characteristics and their behavior in biological models. These studies form a compendium of information that supports the scientific and clinical rationale for development of this carrier for targeted therapeutic interventions.
Collapse
Affiliation(s)
- D M Eckmann
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - R J Composto
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - V R Muzykantov
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|