1
|
Ai H, Pan M, Liu L. Chemical Synthesis of Human Proteoforms and Application in Biomedicine. ACS CENTRAL SCIENCE 2024; 10:1442-1459. [PMID: 39220697 PMCID: PMC11363345 DOI: 10.1021/acscentsci.4c00642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 09/04/2024]
Abstract
Limited understanding of human proteoforms with complex posttranslational modifications and the underlying mechanisms poses a major obstacle to research on human health and disease. This Outlook discusses opportunities and challenges of de novo chemical protein synthesis in human proteoform studies. Our analysis suggests that to develop a comprehensive, robust, and cost-effective methodology for chemical synthesis of various human proteoforms, new chemistries of the following types need to be developed: (1) easy-to-use peptide ligation chemistries allowing more efficient de novo synthesis of protein structural domains, (2) robust temporary structural support strategies for ligation and folding of challenging targets, and (3) efficient transpeptidative protein domain-domain ligation methods for multidomain proteins. Our analysis also indicates that accurate chemical synthesis of human proteoforms can be applied to the following aspects of biomedical research: (1) dissection and reconstitution of the proteoform interaction networks, (2) structural mechanism elucidation and functional analysis of human proteoform complexes, and (3) development and evaluation of drugs targeting human proteoforms. Overall, we suggest that through integrating chemical protein synthesis with in vivo functional analysis, mechanistic biochemistry, and drug development, synthetic chemistry would play a pivotal role in human proteoform research and facilitate the development of precision diagnostics and therapeutics.
Collapse
Affiliation(s)
- Huasong Ai
- New
Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life
Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and
Chemical Biology, Center for Synthetic and Systems Biology, Department
of Chemistry, Tsinghua University, Beijing 100084, China
- Institute
of Translational Medicine, School of Pharmacy, School of Chemistry
and Chemical Engineering, National Center for Translational Medicine
(Shanghai), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Man Pan
- Institute
of Translational Medicine, School of Pharmacy, School of Chemistry
and Chemical Engineering, National Center for Translational Medicine
(Shanghai), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lei Liu
- New
Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life
Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and
Chemical Biology, Center for Synthetic and Systems Biology, Department
of Chemistry, Tsinghua University, Beijing 100084, China
| |
Collapse
|
2
|
Andrianov AK. Delivery of protein therapeutics and vaccines using their multivalent complexes with synthetic polyelectrolytes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 212:235-259. [PMID: 40122646 DOI: 10.1016/bs.pmbts.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Clinical applications of protein and peptide-based therapeutics and vaccines are rapidly expanding. However, the development of promising new product candidates is often hindered by unfavorable pharmacokinetic profiles, which necessitate the implementation of drug delivery systems to improve protein stability and bioavailability. Non-covalent modification of proteins with synthetic polyelectrolytes, which relies on the strength of cooperative multivalent interactions, may offer potential advantages. In contrast to commonly employed covalent conjugation or microencapsulation methodologies, this technology offers dynamic protection of the protein thereby minimizing the loss of its biological activity, enabling "mix-and-match" formulation approaches, reducing manufacturing costs and simplifying regulatory processes. The range of potential life sciences applications ranges from immunopotentiation and vaccine delivery systems to long-circulating stealth biotherapeutics. This review analyses current technology in the context of intended clinical indications and discusses various synthetic and formulation approaches leading to supramolecular complexation. It evaluates dynamic interactions of complexes with constituents of physiological compartments and attempts to identify critical factors that can affect future advancement of this paradigm-shifting protein delivery technology.
Collapse
Affiliation(s)
- Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, United States.
| |
Collapse
|
3
|
Scherger M, Pilger YA, Komforth P, Räder HJ, Nuhn L. Reversible Polymer-Protein Functionalization by Stepwise Introduction of Amine-Reactive, Reductive-Responsive Self-Immolative End Groups onto RAFT-Derived Polymers. ACS Biomater Sci Eng 2024; 10:129-138. [PMID: 36695579 PMCID: PMC10777346 DOI: 10.1021/acsbiomaterials.2c01106] [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: 09/19/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023]
Abstract
Many promising therapeutic protein or peptide drug candidates are rapidly excreted from an organism due to their small size or their inherent immunogenicity. One way to counteract these effects is PEGylation, in which the biopolymer is shielded by synthetic polymers exploiting their stealth properties. However, these modifications are often accompanied by a reduction in the biological function of the protein. By using responsive moieties that bridge the polymer to the protein, a reversible character is provided to this type of conjugation. In this regard, the reductive-responsive nature of disulfides can be exploited via self-immolative structures for reversible linkage to aminic lysine residues and the N-terminus on the protein surface. They enable a traceless release of the intact protein without any further modification and thus preserve the protein's bioactivity. In this study, we demonstrate how this chemistry can be made broadly accessible to RAFT-derived water-soluble polymers like poly(N,N-dimethylacrylamide) (pDMA) as a relevant PEG alternative. A terminal reactive imidazole carbamate with an adjacent self-immolative motif was generated in a gradual manner onto the trithiocarbonate chain transfer moiety of the polymer by first substituting it with a disulfide-bridged alcohol and subsequently converting it into an amine reactive imidazole carbamate. Successful synthesis and complete characterization were demonstrated by NMR, size exclusion chromatography, and mass spectrometry. Finally, two model proteins, lysozyme and a therapeutically relevant nanobody, were functionalized with the generated polymer, which was found to be fully reversible under reductive conditions in the presence of free thiols. This strategy has the potential to extend the generation of reversible reductive-responsive polymer-protein hybrids to the broad field of available functional RAFT-derived polymers.
Collapse
Affiliation(s)
- Maximilian Scherger
- Max
Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Yannick A. Pilger
- Max
Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
- Chair
of Macromolecular Chemistry, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg 97070, Germany
| | - Patric Komforth
- Max
Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Hans-Joachim Räder
- Max
Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Lutz Nuhn
- Max
Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
- Chair
of Macromolecular Chemistry, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg 97070, Germany
| |
Collapse
|
4
|
Yu Q, Bai L, Jiang X. Disulfide Click Reaction for Stapling of S-terminal Peptides. Angew Chem Int Ed Engl 2023; 62:e202314379. [PMID: 37950389 DOI: 10.1002/anie.202314379] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 11/12/2023]
Abstract
A disulfide click strategy is disclosed for stapling to enhance the metabolic stability and cellular permeability of therapeutic peptides. A 17-membered library of stapling reagents with adjustable lengths and angles was established for rapid double/triple click reactions, bridging S-terminal peptides from 3 to 18 amino acid residues to provide 18- to 48-membered macrocyclic peptides under biocompatible conditions. The constrained peptides exhibited enhanced anti-HCT-116 activity with a locked α-helical conformation (IC50 =6.81 μM vs. biological incompetence for acyclic linear peptides), which could be unstapled for rehabilitation of the native peptides under the assistance of tris(2-carboxyethyl)phosphine (TCEP). This protocol assembles linear peptides into cyclic peptides controllably to retain the diverse three-dimensional conformations, enabling their cellular uptake followed by release of the disulfides for peptide delivery.
Collapse
Affiliation(s)
- Qing Yu
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, P. R. China
| | - Leiyang Bai
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, P. R. China
| | - Xuefeng Jiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, P. R. China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, P. R. China
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, P. R. China
| |
Collapse
|
5
|
Andrianov AK. Noncovalent PEGylation of protein and peptide therapeutics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1897. [PMID: 37138514 DOI: 10.1002/wnan.1897] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/15/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023]
Abstract
Clinical applications of protein therapeutics-an advanced generation of drugs characterized by high biological specificity-are rapidly expanding. However, their development is often impeded by unfavorable pharmacokinetic profiles and largely relies on the use of drug delivery systems to prolong their in vivo half-life and suppress undesirable immunogenicity. Although a commercially established PEGylation technology based on protein conjugation with poly(ethylene glycol) (PEG)-protective steric shield resolves some of the challenges, the search for alternatives continues. Noncovalent PEGylation, which mainly relies on multivalent (cooperative) interactions and high affinity (host-guest) complexes formed between protein and PEG offers a number of potential advantages. Among them are dynamic or reversible protection of the protein with minimal loss of biological activity, drastically lower manufacturing costs, "mix-and-match" formulations approaches, and expanded scope of PEGylation targets. While a great number of innovative chemical approaches have been proposed in recent years, the ability to effectively control the stability of noncovalently assembled protein-PEG complexes under physiological conditions presents a serious challenge for the commercial development of the technology. In an attempt to identify critical factors affecting pharmacological behavior of noncovalently linked complexes, this Review follows a hierarchical analysis of various experimental techniques and resulting supramolecular architectures. The importance of in vivo administration routes, degradation patterns of PEGylating agents, and a multitude of potential exchange reactions with constituents of physiological compartments are highlighted. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Alexander K Andrianov
- Institute of Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, USA
| |
Collapse
|
6
|
Jeong SH, Cheong S, Kim TY, Choi H, Hahn SK. Supramolecular Hydrogels for Precisely Controlled Antimicrobial Peptide Delivery for Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2023; 15:16471-16481. [PMID: 36943445 DOI: 10.1021/acsami.3c00191] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Diabetic wound patients are often exposed to bacterial infections with delayed healing process due to hyperglycemia in the damaged skin tissue. Antimicrobial peptides (AMPs) have been investigated for the treatment of infection-induced diabetic wounds, but their low stability and toxicity have limited their further applications to diabetic chronic wound healing. Here, we developed a precisely controlled AMP-releasing injectable hydrogel platform, which could respond to infection-related materials of matrix metalloproteinases (MMPs) and reactive oxygen species (ROS). The injectable supramolecular hydrogel was prepared by the simple mixing of hyaluronic acid modified with cyclodextrin (HA-CD) and adamantane (Ad-HA). Ad-HA was conjugated with AMP via the cyclic peptide linker composed of MMP and ROS cleavable sequence (Ad-HA-AMP). Remarkably, only when the AMP-tethered hydrogel was exposed to both MMP and ROS simultaneously, AMP was released from the hydrogel, enabling the controlled release of AMP without causing cytotoxicity. In addition, we confirmed the enhanced serum stability of the Ad-HA-AMP conjugate. The antimicrobial activity of Ad-HA-AMP was maintained much longer than that of the native AMP. Finally, we could demonstrate the greatly improved wound-healing effect of AMP-tethered hydrogels with enhanced safety for the treatment of infection-induced diabetic chronic wounds. Taken together, we successfully demonstrated the feasibility of sHG-AMP for diabetic chronic wound healing.
Collapse
Affiliation(s)
- Sang Hoon Jeong
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 790-784, Korea
| | - Sunah Cheong
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 790-784, Korea
| | - Tae Yeon Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 790-784, Korea
| | - Hyunsik Choi
- PHI Biomed Co., 168 Yeoksam-ro, Gangnam-gu, Seoul 06248, Korea
| | - Sei Kwang Hahn
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 790-784, Korea
- PHI Biomed Co., 168 Yeoksam-ro, Gangnam-gu, Seoul 06248, Korea
| |
Collapse
|
7
|
Proteins and their functionalization for finding therapeutic avenues in cancer: Current status and future prospective. Biochim Biophys Acta Rev Cancer 2023; 1878:188862. [PMID: 36791920 DOI: 10.1016/j.bbcan.2023.188862] [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: 06/24/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/15/2023]
Abstract
Despite the remarkable advancement in the health care sector, cancer remains the second most fatal disease globally. The existing conventional cancer treatments primarily include chemotherapy, which has been associated with little to severe side effects, and radiotherapy, which is usually expensive. To overcome these problems, target-specific nanocarriers have been explored for delivering chemo drugs. However, recent reports on using a few proteins having anticancer activity and further use of them as drug carriers have generated tremendous attention for furthering the research towards cancer therapy. Biomolecules, especially proteins, have emerged as suitable alternatives in cancer treatment due to multiple favourable properties including biocompatibility, biodegradability, and structural flexibility for easy surface functionalization. Several in vitro and in vivo studies have reported that various proteins derived from animal, plant, and bacterial species, demonstrated strong cytotoxic and antiproliferative properties against malignant cells in native and their different structural conformations. Moreover, surface tunable properties of these proteins help to bind a range of anticancer drugs and target ligands, thus making them efficient delivery agents in cancer therapy. Here, we discuss various proteins obtained from common exogenous sources and how they transform into effective anticancer agents. We also comprehensively discuss the tumor-killing mechanisms of different dietary proteins such as bovine α-lactalbumin, hen egg-white lysozyme, and their conjugates. We also articulate how protein nanostructures can be used as carriers for delivering cancer drugs and theranostics, and strategies to be adopted for improving their in vivo delivery and targeting. We further discuss the FDA-approved protein-based anticancer formulations along with those in different phases of clinical trials.
Collapse
|
8
|
Zeng Z, Zhu J, Deng X, Chen H, Jin Y, Miclet E, Alezra V, Wan Y. Customized Reversible Stapling for Selective Delivery of Bioactive Peptides. J Am Chem Soc 2022; 144:23614-23621. [PMID: 36530144 DOI: 10.1021/jacs.2c10949] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We have developed a new concept for reversible peptide stapling that involves macrocyclization between two amino groups and decyclization promoted via dual 1,4-elimination. Depending on the trigger moiety, this strategy could be employed to selectively deliver peptides to either intracellular or extracellular targets. As a proof of concept, a peptide inhibitor targeting a lysine-specific demethylase 1 (LSD1) was temporarily cyclized to enhance its stability and ability to cross the cell membrane. Once inside the cells, the biologically active linear peptide was released under reducing environment. Moreover, we have developed reversibly stapled peptides using antimicrobial peptides (RStAMPs) whose bioactive helical conformation can be temporarily destabilized by stapling the peptide backbone. The resulting helix-distorted RStAMPs are nontoxic and highly resistant to protease hydrolysis, while at the infection site, RStAMPs can be rapidly activated by the overproduced H2O2 through the dual 1,4-elimination. The latter restored the helical structure of the native peptide and its antimicrobial activity. This work illustrates a highly valuable macrocyclization strategy for the peptide community and should greatly benefit the field of peptide delivery.
Collapse
Affiliation(s)
- Zizhen Zeng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, P. R. China
| | - Jibao Zhu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, P. R. China
| | - Xiaoyu Deng
- Minist Educ, Key Lab Modern Preparat TCM, Jiangxi University of Chinese Medicine, Nanchang 330006, P. R. China
| | - Huanwen Chen
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, P. R. China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, P. R. China
| | - Emeric Miclet
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, 4 Place Jussieu, Cedex 05, Paris 75252, France
| | - Valérie Alezra
- Laboratoire de Méthodologie, Synthèse et Molécules Thérapeutiques, ICMMO, Université Paris-Saclay, Paris 91400, Orsay, France
| | - Yang Wan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, P. R. China
| |
Collapse
|
9
|
Kaupbayeva B, Murata H, Rule GS, Matyjaszewski K, Russell AJ. Rational Control of Protein-Protein Interactions with Protein-ATRP-Generated Protease-Sensitive Polymer Cages. Biomacromolecules 2022; 23:3831-3846. [PMID: 35984406 DOI: 10.1021/acs.biomac.2c00679] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protease-protease interactions lie at the heart of the biological cascades that provide rapid molecular responses to living systems. Blood clotting cascades, apoptosis signaling networks, bacterial infection, and virus trafficking have all evolved to be activated and sustained by protease-protease interactions. Biomimetic strategies designed to target drugs to specific locations have generated proprotein drugs that can be activated by proteolytic cleavage to release native protein. We have previously demonstrated that the modification of enzymes with a custom-designed comb-shaped polymer nanoarmor can shield the enzyme surface and eliminate almost all protein-protein interactions. We now describe the synthesis and characterization of protease-sensitive comb-shaped nanoarmor cages using poly(ethylene glycol) [Sundy, J. S. Arthritis Rheum. 2008, 58(9), 2882-2891]methacrylate macromonomers where the PEG tines of the comb are connected to the backbone of the growing polymer chain by peptide linkers. Protease-induced cleavage of the tines of the comb releases a polymer-modified protein that can once again participate in protein-protein interactions. Atom transfer radical polymerization (ATRP) was used to copolymerize the macromonomer and carboxybetaine methacrylate from initiator-labeled chymotrypsin and trypsin enzymes, yielding proprotease conjugates that retained activity toward small peptide substrates but prevented activity against proteins. Native proteases triggered the release of the PEG side chains from the polymer backbone within 20 min, thereby increasing the activity of the conjugate toward larger protein substrates by 100%. Biomimetic cascade initiation of nanoarmored protease-sensitive protein-polymer conjugates may open the door to a new class of responsive targeted therapies.
Collapse
Affiliation(s)
- Bibifatima Kaupbayeva
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States.,Center for Polymer-Based Protein Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States.,National Laboratory Astana, Nazarbayev University, Nur-Sultan City 010000, Kazakhstan
| | - Hironobu Murata
- Center for Polymer-Based Protein Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States.,Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Gordon S Rule
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States.,Center for Polymer-Based Protein Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Krzysztof Matyjaszewski
- Center for Polymer-Based Protein Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States.,Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Alan J Russell
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States.,Center for Polymer-Based Protein Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States.,Department of Chemical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States.,Amgen, 1 Amgen Center Drive, Thousand Oaks, California 91320, United States
| |
Collapse
|
10
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
11
|
Wright TA, Rahman MS, Bennett C, Johnson MR, Fischesser H, Ram N, Tyler A, Page RC, Konkolewicz D. Hydrolytically Stable Maleimide-End-Functionalized Polymers for Site-Specific Protein Conjugation. Bioconjug Chem 2021; 32:2447-2456. [PMID: 34730954 PMCID: PMC9099401 DOI: 10.1021/acs.bioconjchem.1c00487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Site-specific conjugation to cysteines of proteins often uses ester groups to link maleimide or alkene groups to polymers. However, the ester group is susceptible to hydrolysis, potentially losing the benefits gained through bioconjugation. Here, we present a simple conjugation strategy that utilizes the amide bond stability of traditional 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide/N-hydroxysuccinimide coupling while introducing site specificity. Hydrolytically stable maleimide-end-functionalized polymers for site-specific conjugation to free cysteines of proteins were synthesized using reversible addition-fragmentation chain-transfer (RAFT) polymerization. The alpha terminus of the polymers was amidated with a furan-protected aminoethyl maleimide using carbodiimide-based chemistry. Finally, the maleimide was exposed by a retro Diels-Alder reaction to yield the maleimide group, allowing for thiol-maleimide click chemistry for bioconjugation. A thermophilic cellulase from Fervidobacterium nodosum (FnCel5a) was conjugated using various strategies, including random 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC)/N-hydroxysuccinimide (NHS) coupling, site-specific hydroxyethyl maleimide (HEMI) end-functionalized coupling, hydroxyethyl acrylate (HEA) end-functionalized coupling, and amidoethyl maleimide (AEMI) end-functionalized coupling. Only the polymers conjugated by EDC and AEMI remained conjugated a week after attachment. This indicates that hydrolytically stable amide-based maleimides are an important bioconjugation strategy for conjugates that require long-term stability, while esters are better suited for systems that require debonding of polymers over time.
Collapse
Affiliation(s)
- Thaiesha A Wright
- Department of Chemistry and Biochemistry, Miami University, 651 East High Street, Oxford, Ohio 45056, United States
| | - Monica Sharfin Rahman
- Department of Chemistry and Biochemistry, Miami University, 651 East High Street, Oxford, Ohio 45056, United States
| | - Camaryn Bennett
- Department of Chemistry and Biochemistry, Miami University, 651 East High Street, Oxford, Ohio 45056, United States
| | - Madolynn R Johnson
- Department of Chemistry and Biochemistry, Miami University, 651 East High Street, Oxford, Ohio 45056, United States
| | - Henry Fischesser
- Department of Chemistry and Biochemistry, Miami University, 651 East High Street, Oxford, Ohio 45056, United States
| | - Natasha Ram
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, 4202 East Fowler Ave, Tampa, Florida 33620, United States
| | - Amoni Tyler
- Department of Agricultural and Life Sciences, Central State University, 1400 Brush Row Road, Wilberforce, Ohio 45384, United States
| | - Richard C Page
- Department of Chemistry and Biochemistry, Miami University, 651 East High Street, Oxford, Ohio 45056, United States
| | - Dominik Konkolewicz
- Department of Chemistry and Biochemistry, Miami University, 651 East High Street, Oxford, Ohio 45056, United States
| |
Collapse
|
12
|
Docetaxel encapsulation in nanoscale assembly micelles of folate-PEG-docetaxel conjugates for targeted fighting against metastatic breast cancer in vitro and in vivo. Int J Pharm 2021; 605:120822. [PMID: 34182039 DOI: 10.1016/j.ijpharm.2021.120822] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 01/17/2023]
Abstract
Due to the high frequency and mortality of breast cancer, developing efficient targeted drug delivery systems for frightening against this malignancy is among cancer research priorities. The aim of this study was to synthesize a targeted micellar formulation of docetaxel (DTX) using DTX, folic acid (FA) and polyethylene glycol (PEG) conjugates as building blocks. In the current study, two therapeutic polymers consisting of FA-PEG-DTX and PEG-DTX conjugates were synthesized and implemented to form folate-targeted and non-targeted micelles. Dissipative particle dynamics (DPD) method was used to simulate the behavior of the nanoparticle. The anti-cancer drug, DTX was loaded in to the micelles via solvent switching method in order to increase its solubility and stability. The cytotoxicity of the targeted and non-targeted formulations was evaluated against 4T1 and CHO cell lines. In vivo therapeutic efficiency was studied using ectopic tumor model of metastatic breast cancer, 4T1, in Female BALB/c mice. The successful synthesis of therapeutic polymers, FA-PEG-DTX and PEG-DTX were confirmed implementing 1HNMR spectral analysis. The size of DTX-loaded non-targeted and targeted micelles were 176.3 ± 8.3 and 181 ± 10.1 nm with PDI of 0.23 and 0.17, respectively. Loading efficiencies of DTX in non-targeted and targeted micelles were obtained to be 85% and 82%, respectively. In vitro release study at pH = 7.4 and pH = 5.4 showed a controlled and continuous drug release for both formulations, that was faster at pH = 5.4 (100% drug release within 120 h) than at pH = 7.4 (80% drug release within 150 h). The targeted formulation showed a significant higher cytotoxicity against 4T1 breast cancer cells (high expression of folate receptor) within the range of 12.5 to 200 μg/mL in comparison with no-targeted one. However, there was no significant difference between the cytotoxicity of the targeted and non-targeted formulations against CHO cell line as low-expressed cell line. In accordance with in vitro investigation, in vivo studies verified the ideal anti-tumor efficacy of the targeted formulation compared to Taxotere and non-targeted formulation. Based on the obtained data, FA-targeted DTX-loaded nano-micelles significantly increased the therapeutic efficacy of DTX and therefore can be considered as a new potent platform for breast cancer chemotherapy.
Collapse
|
13
|
Redox-responsive prodrug for improving oral bioavailability of paclitaxel through bile acid transporter-mediated pathway. Int J Pharm 2021; 600:120496. [PMID: 33746013 DOI: 10.1016/j.ijpharm.2021.120496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/23/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Most anticancer drugs are not orally bioavailable due to their undesirable physicochemical properties and inherent physiological barriers. In this study, a polymeric prodrug strategy was presented to enhance the oral bioavailability of BCS class IV drugs using paclitaxel (PTX) as the model drug. PTX was covalently conjugated with cholic acid-functionalized PEG by a redox-sensitive disulfide bond. Cholic acid-functionalized PEGylated PTX (CPP) achieved remarkably improved PTX solubility (>30,000-fold), as well as favorable stability under the physiological environment and controlled drug release in the tumor. Meanwhile, CPP could self-assemble into nanoparticles with an average size of 56.18 ± 2.06 nm and drug loading up to 17.6% (w/w). Then, permeability study on Caco-2 cell monolayers demonstrated that CPP obtained an approximately 4-fold increase by apical sodium-dependent bile acid transporter (ASBT) mediated transport, compared with Taxol®. Pharmacokinetic studies carried out in rats confirmed that the oral bioavailability of CPP was 10-fold higher than that of Taxol®. Finally, significant improvement in the antitumor efficacy of CPP against breast cancer was confirmed on MDA-MB-231 cells. In summary, this prodrug-based cascade strategy offers new ways for chemotherapeutic drugs whose oral delivery is limited by solubility and permeability, also endows drugs with the capacity of tumor-specific release.
Collapse
|
14
|
Bergmann R, Chollet C, Els-Heindl S, Ullrich M, Berndt N, Pietzsch J, Máthé D, Bachmann M, Beck-Sickinger AG. Development of a ghrelin receptor inverse agonist for positron emission tomography. Oncotarget 2021; 12:450-474. [PMID: 33747360 PMCID: PMC7939532 DOI: 10.18632/oncotarget.27895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Imaging of Ghrelin receptors in vivo provides unique potential to gain deeper understanding on Ghrelin and its receptors in health and disease, in particular, in cancer. Ghrelin, an octanoylated 28-mer peptide hormone activates the constitutively active growth hormone secretagogue receptor type 1a (GHS-R1a) with nanomolar activity. We developed novel compounds, derived from the potent inverse agonist K-(D-1-Nal)-FwLL-NH2 but structurally varied by lysine conjugation with 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid (NODAGA), palmitic acid and/or diethylene glycol (PEG2) to allow radiolabeling and improve pharmacokinetics, respectively. All compounds were tested for receptor binding, potency and efficacy in vitro, for biodistribution and -kinetics in rats and in preclinical prostate cancer models on mice. Radiolabeling with Cu-64 and Ga-68 was successfully achieved. The Cu-64- or Ga-68-NODAGA-NH-K-K-(D-1-NaI)-F-w-L-L-NH2 radiotracer were specifically accumulated by the GHS-R1a in xenotransplanted human prostate tumor models (PC-3, DU-145) in mice. The tumors were clearly delineated by PET. The radiotracer uptake was also partially blocked by K-(D-1-Nal)-FwLL-NH2 in stomach and thyroid. The presence of the GHS-R1a was also confirmed by immunohistology. In the arterial rat blood plasma, only the original compounds were found. The Cu-64 or Ga-68-NODAGA-NH-K-K-(D-1-NaI)-F-w-L-L-NH2 radiolabeled inverse agonists turned out to be potent and safe. Due to their easy synthesis, high affinity, medium potency, metabolic stability, and the suitable pharmacokinetic profiles, they are excellent tools for imaging and quantitation of GHS-R1a expression in normal and cancer tissues by PET. These compounds can be used as novel biomarkers of the Ghrelin system in precision medicine.
Collapse
Affiliation(s)
- Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary.,These authors contributed equally to this work
| | - Constance Chollet
- Institute of Biochemistry, Faculty of Life Sciences, Universität Leipzig, Leipzig, Germany.,These authors contributed equally to this work
| | - Sylvia Els-Heindl
- Institute of Biochemistry, Faculty of Life Sciences, Universität Leipzig, Leipzig, Germany
| | - Martin Ullrich
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Nicole Berndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Tumor Immunology, University Cancer Center, Carl Gustav Carus Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases, Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | | |
Collapse
|
15
|
Marques MMB, Santos AS. Pd-Catalyzed Functionalization of Aryl Amines on a Soluble Polymer Support. Synlett 2020. [DOI: 10.1055/s-0040-1707261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AbstractHerein we report the use of a soluble polymer support PEG-2000 on Pd-catalyzed reactions to improve the functionalization of aromatic amines and the synthesis of N-heterocycles. Compatibility of metal-catalyzed reactions for assembling privileged structures such as functionalized anilines were studied. PEG-supported anilines were found to be suitable substrates for Pd-catalyzed N-arylation, Sonogashira and Heck reactions. PEGylated substrates were prepared in yields up to 94%. This work consists on a proof of concept on the use of PEGylated anilines on Pd-catalyzed cross-coupling reactions. Indole core was attained in 82% and 62% yields, via two different routes.
Collapse
|
16
|
Nieto-Orellana A, Li H, Rosiere R, Wauthoz N, Williams H, Monteiro CJ, Bosquillon C, Childerhouse N, Keegan G, Coghlan D, Mantovani G, Stolnik S. Targeted PEG-poly(glutamic acid) complexes for inhalation protein delivery to the lung. J Control Release 2019; 316:250-262. [PMID: 31678655 DOI: 10.1016/j.jconrel.2019.10.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/02/2019] [Accepted: 10/06/2019] [Indexed: 12/18/2022]
Abstract
Pulmonary delivery is increasingly seen as an attractive, non-invasive route for the delivery of forthcoming protein therapeutics. In this context, here we describe protein complexes with a new 'complexing excipient' - vitamin B12-targeted poly(ethylene glycol)-block-poly(glutamic acid) copolymers. These form complexes in sub-200nm size with a model protein, suitable for cellular targeting and intracellular delivery. Initially we confirmed expression of vitamin B12-internalization receptor (CD320) by Calu-3 cells of the in vitro lung epithelial model used, and demonstrated enhanced B12 receptor-mediated cellular internalization of B12-targeted complexes, relative to non-targeted counterparts or protein alone. To develop an inhalation formulation, the protein complexes were spray dried adopting a standard protocol into powders with aerodynamic diameter within the suitable range for lower airway deposition. The cellular internalization of targeted complexes from dry powders applied directly to Calu-3 model was found to be 2-3 fold higher compared to non-targeted complexes. The copolymer complexes show no complement activation, and in vivo lung tolerance studies demonstrated that repeated administration of formulated dry powders over a 3 week period in healthy BALB/c mice induced no significant toxicity or indications of lung inflammation, as assessed by cell population count and quantification of IL-1β, IL-6, and TNF-α pro-inflammatory markers. Importantly, the in vivo data appear to suggest that B12-targeted polymer complexes administered as dry powder enhance lung retention of their protein payload, relative to protein alone and non-targeted counterparts. Taken together, our data illustrate the potential developability of novel B12-targeted poly(ethylene glycol)-poly(glutamic acid) copolymers as excipients suitable to be formulated into a dry powder product for the inhalation delivery of proteins, with no significant lung toxicity, and with enhanced protein retention at their in vivo target tissue.
Collapse
Affiliation(s)
- A Nieto-Orellana
- Molecular Therapeutics and Formulation Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - H Li
- Molecular Therapeutics and Formulation Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - R Rosiere
- Laboratory of Pharmaceutics and Biopharmaceutics (ULBGAL), Université Libre de Bruxelles, Bruxelles, BE, Belgium
| | - N Wauthoz
- Laboratory of Pharmaceutics and Biopharmaceutics (ULBGAL), Université Libre de Bruxelles, Bruxelles, BE, Belgium
| | - H Williams
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - C J Monteiro
- Molecular Therapeutics and Formulation Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - C Bosquillon
- Molecular Therapeutics and Formulation Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | - G Keegan
- Vectura Group plc, Chippenhafm, UK
| | | | - G Mantovani
- Molecular Therapeutics and Formulation Division, School of Pharmacy, University of Nottingham, Nottingham, UK.
| | - S Stolnik
- Molecular Therapeutics and Formulation Division, School of Pharmacy, University of Nottingham, Nottingham, UK.
| |
Collapse
|
17
|
Strategies for the production of long-acting therapeutics and efficient drug delivery for cancer treatment. Biomed Pharmacother 2019; 113:108750. [DOI: 10.1016/j.biopha.2019.108750] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 11/21/2022] Open
|
18
|
Abstract
Introduction: PEGylation is a well-established technology for improving the therapeutic value of drugs by attaching polyethylene glycol (PEG). The first PEGylated enzyme products appeared on the market in the early 1990s; currently, more than 18 PEGylated products have been approved by Food and Drug Administration, which encompass various classes of drug molecules, such as enzymes, interferons, granulocyte colony-stimulating factors, hormones, antibody fragments, coagulation factors, oligonucleotide aptamers, synthetic peptides, and small organic molecules. Areas covered: While PEGylated products mainly comprise biologic drugs, such as recombinant proteins and enzymes, non-biologic drugs have recently emerged as a target for PEGylation. This review focuses on the recent development of PEGylated non-biologic drugs, such as small organic molecules, synthetic peptides, and aptamers. Expert opinion: Several PEGylated versions of anti-cancer drugs, opioid agonists, glucagon-like peptide-1 receptor agonists, and oligonucleotide aptamers are in active development stage, and it is likely that they will have a dramatic impact on the market. Although some safety concerns about PEG in clinical trials have been recently issued, PEGylation is still a commercially attractive proposition as a half-life extension technology for long-acting drug development.
Collapse
Affiliation(s)
- Eun Ji Park
- a College of Pharmacy , Chung-Ang University , Seoul , Republic of Korea.,b D&D Pharmatech , Seongnam , Republic of Korea
| | - Jiyoung Choi
- a College of Pharmacy , Chung-Ang University , Seoul , Republic of Korea
| | - Kang Choon Lee
- b D&D Pharmatech , Seongnam , Republic of Korea.,c College of Pharmacy , SungKyunKwan University , Suwon , Republic of Korea
| | - Dong Hee Na
- a College of Pharmacy , Chung-Ang University , Seoul , Republic of Korea
| |
Collapse
|
19
|
Hildebrand N, Wei G, Köppen S, Colombi Ciacchi L. Simulated and experimental force spectroscopy of lysozyme on silica. Phys Chem Chem Phys 2018; 20:19595-19605. [PMID: 30009290 DOI: 10.1039/c8cp03747g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The force spectra of proteins detaching from oxide surfaces measured by atomic force microscopy (AFM) often present complex patterns of peaks, which are difficult to correlate with individual bond-breaking events at the atomic scale. In this work we rationalize experimental AFM force spectra of hen-egg-white lysozyme detaching from silica by means of all-atom steered molecular dynamics (SMD) simulations. In particular, we demonstrate that the native tertiary structure of lysozyme is preserved if, and only if, its four intramolecular disulfide bridges are intact. Otherwise, the protein pulled off the surface undergoes severe unfolding, which is well captured by SMD simulations in explicit solvent. Implicit solvent simulations, on the contrary, wrongly predict protein unfolding even in the presence of S-S bridges, due to the lack of additional structural stabilization provided by the water's hydrogen-bond network within and surrounding the protein. On the basis of our combined experimental and theoretical findings, we infer that the rugged force spectra characteristic of lysozyme/silica interfaces are not due to the successive breaking of internal disulfide bonds leading to partial unfolding events. Rather, they reflect the detachment of several molecules bound to the same AFM tip, each anchored to the surface via multiple hydrogen and ionic bonds.
Collapse
Affiliation(s)
- Nils Hildebrand
- Hybrid Materials Interfaces Group, Faculty Production Engineering, Bremen Center for Computational Materials Science, University of Bremen, Am Fallturm 1, 28359 Bremen, Germany.
| | | | | | | |
Collapse
|
20
|
Bioresponsive release of insulin-like growth factor-I from its PEGylated conjugate. J Control Release 2018; 279:17-28. [DOI: 10.1016/j.jconrel.2018.04.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 02/06/2023]
|
21
|
Braun AC, Gutmann M, Lühmann T, Meinel L. Bioorthogonal strategies for site-directed decoration of biomaterials with therapeutic proteins. J Control Release 2018; 273:68-85. [PMID: 29360478 DOI: 10.1016/j.jconrel.2018.01.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 01/04/2023]
Abstract
Emerging strategies targeting site-specific protein modifications allow for unprecedented selectivity, fast kinetics and mild reaction conditions with high yield. These advances open exciting novel possibilities for the effective bioorthogonal decoration of biomaterials with therapeutic proteins. Site-specificity is particularly important to the therapeutics' end and translated by targeting specific functional groups or introducing new functional groups into the therapeutic at predefined positions. Biomimetic strategies are designed for modification of therapeutics emulating enzymatic strategies found in Nature. These strategies are suitable for a diverse range of applications - not only for protein-polymer conjugation, particle decoration and surface immobilization, but also for the decoration of complex biomaterials and the synthesis of bioresponsive drug delivery systems. This article reviews latest chemical and enzymatic strategies for the biorthogonal decoration of biomaterials with therapeutic proteins and inter-positioned linker structures. Finally, the numerous reports at the interface of biomaterials, linkers, and therapeutic protein decoration are integrated into practical advice for design considerations intended to support the selection of productive ligation strategies.
Collapse
Affiliation(s)
- Alexandra C Braun
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Marcus Gutmann
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Tessa Lühmann
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany.
| |
Collapse
|
22
|
Hagiwara K, Kurihara K, Honma M, Yamamoto J, Shinohara F. PEG-modification on the endo-position of an antisense oligonucleotide increases tumor accumulation via the EPR effect. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:448-459. [PMID: 29318941 DOI: 10.1080/09205063.2017.1422853] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Nucleic acid medicine is the next-generation therapeutic modality for refractory diseases with its unique mode of action as an alternative to traditional therapies. A nucleic acid delivery system targeted to liver was validated clinically; however, the delivery system of nucleic acids targeting solid tumors following systemic administration is not efficient enough for clinical use. In this study, we first utilized an antisense oligonucleotide (ASO) and polyethylene glycol (PEG) in one-to-one conjugation (PEG-ASO) at the endo-position of the ASO (endo-PEG-ASO). The effects of ASO modification position, PEG structure and molecular weight, and PEG-ASO tumor accumulation were evaluated in vivo. The endo-PEG-ASO showed prolonged pharmacokinetics and enhanced tumor accumulation compared with the conventional ASO and the PEG-ASO modified at the ASO exo-position (exo-PEG-ASO), indicating that the modification position of PEG is crucial for targeting tumors. We also observed that the endo-PEG-ASO indicated possibility of enhanced permeability inside the tumor. Further research is needed to optimize the linker in the endo-PEG-ASO for clinical application as a novel and promising therapeutic format for targeting solid tumors.
Collapse
Affiliation(s)
- Kenji Hagiwara
- a Innovative Technology Laboratories, Research Functions Unit, R&D Division , Kyowa Hakko Kirin Co., Ltd , Tokyo , Japan
| | - Kana Kurihara
- b Research Core Function Laboratories, Research Functions Unit, R&D Division , Kyowa Hakko Kirin Co., Ltd , Tokyo , Japan
| | - Masakazu Honma
- a Innovative Technology Laboratories, Research Functions Unit, R&D Division , Kyowa Hakko Kirin Co., Ltd , Tokyo , Japan
| | - Junichiro Yamamoto
- a Innovative Technology Laboratories, Research Functions Unit, R&D Division , Kyowa Hakko Kirin Co., Ltd , Tokyo , Japan
| | - Fumikazu Shinohara
- a Innovative Technology Laboratories, Research Functions Unit, R&D Division , Kyowa Hakko Kirin Co., Ltd , Tokyo , Japan
| |
Collapse
|
23
|
Ahmadi Z, Verma G, Jha D, Gautam HK, Kumar P. Evaluation of antimicrobial activity and cytotoxicity of pegylated aminoglycosides. J BIOACT COMPAT POL 2017. [DOI: 10.1177/0883911517739318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Zeba Ahmadi
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Geeta Verma
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Diksha Jha
- Microbial Technology Laboratory, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Hemant Kumar Gautam
- Microbial Technology Laboratory, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| |
Collapse
|
24
|
Perspectives on dendritic architectures and their biological applications: From core to cell. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 173:61-83. [PMID: 28564631 DOI: 10.1016/j.jphotobiol.2017.05.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/15/2017] [Accepted: 05/18/2017] [Indexed: 12/24/2022]
Abstract
The challenges of medicine today include the increasing stipulation for sensitive and effective systems that can improve the pathological responses with a simultaneous reduction in accumulation and drug side effects. The demand can be fulfilled through the advancements in nanomedicine that includes nanostructures and nanodevices for diagnosing, treating, and prevention of various diseases. In this respect, the nanoscience provides various novel techniques with carriers such as micelles, dendrimers, particles and vesicles for the transportation of active moieties. Further, an efficient way to improve these systems is through stimuli a responsive system that utilizes supramolecular hyperbranched structures to meet the above criteria. The stimuli-responsive dendritic architectures exhibit spatial, temporal, convenient, effective, safety and controlled drug release in response to specific trigger through electrostatic interactions plus π stacking. The stimuli-responsive systems are capable of sequestering the drug molecules underneath a predefined set of conditions and discharge them in a different environment through either exogenous or endogenous stimulus. The incorporation of photoresponsive moieties at various components of dendrimer such as core, branches or at the peripheral end exaggerates its significance in various allied fields of nanotechnology which includes sensors, photoswitch, electronic widgets and in drug delivery systems. This is due to the light instigated geometrical modifications at the core or at the surface molecules which generates huge conformational changes throughout the hyperbranched structure. Further, numerous synthetic methodologies have been investigated for utilization of dendrimers in therapeutic drug delivery and its applicability towards stimuli responsive systems such as photo-instigated, thermal-instigated, and pH-instigated hyperbranched structures and their advancement in the field of nanomedicine. This paper highlights the fascinating theoretical advances and principal mechanisms of dendrimer synthesis and their ability to capture light that strengthens its applicability from radiant energy to medical photonics.
Collapse
|
25
|
Ma S, We L, Yang H, Deng S, M. Jevnikar A. Emerging technologies to achieve oral delivery of GLP-1 and GLP-1 analogs for treatment of type 2 diabetes mellitus (T2DM). CANADIAN JOURNAL OF BIOTECHNOLOGY 2017. [DOI: 10.24870/cjb.2017-000107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
26
|
Gong Y, Andina D, Nahar S, Leroux JC, Gauthier MA. Releasable and traceless PEGylation of arginine-rich antimicrobial peptides. Chem Sci 2017; 8:4082-4086. [PMID: 30155213 PMCID: PMC6094173 DOI: 10.1039/c7sc00770a] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/21/2017] [Indexed: 02/01/2023] Open
Abstract
This study reports a strategy to temporarily mask arginine residues within antimicrobial peptides (AMPs) with methoxy poly(ethylene glycol) (mPEG). PEGylation protects AMPs from serum proteases, and can be released at a pharmaceutically-relevant rate. Fully active and unmodified (i.e., native) AMPs are released with time.
Arginine-rich antimicrobial peptides (AMPs) are emerging therapeutics of interest. However, their applicability is limited by their short circulation half-life, caused in part by their small size and digestion by blood proteases. This study reports a strategy to temporarily mask arginine residues within AMPs with methoxy poly(ethylene glycol). Based on the reagent used, release of AMPs occurred in hours to days in a completely traceless fashion. In vitro, conjugates were insensitive to serum proteases, and released native AMP with full in vitro bioactivity. This strategy is thus highly relevant and should be adaptable to the entire family of arginine-rich AMPs. It may potentially be used to improve AMP-therapies by providing a more steady concentration of AMP in the blood after a single injection, avoiding toxic effects at high AMP doses, and reducing the number of doses required over the treatment duration.
Collapse
Affiliation(s)
- Y Gong
- Institute of Pharmaceutical Sciences , Department of Chemistry and Applied Biosciences , Swiss Federal Institute of Technology Zurich (ETHZ) , Zurich 8093 , Switzerland
| | - D Andina
- Institute of Pharmaceutical Sciences , Department of Chemistry and Applied Biosciences , Swiss Federal Institute of Technology Zurich (ETHZ) , Zurich 8093 , Switzerland
| | - S Nahar
- EMT Research Center , Institut National de la Recherche Scientifique (INRS) , Varennes J3X 1S2 , Canada .
| | - J-C Leroux
- Institute of Pharmaceutical Sciences , Department of Chemistry and Applied Biosciences , Swiss Federal Institute of Technology Zurich (ETHZ) , Zurich 8093 , Switzerland
| | - M A Gauthier
- EMT Research Center , Institut National de la Recherche Scientifique (INRS) , Varennes J3X 1S2 , Canada .
| |
Collapse
|
27
|
Turecek PL, Bossard MJ, Schoetens F, Ivens IA. PEGylation of Biopharmaceuticals: A Review of Chemistry and Nonclinical Safety Information of Approved Drugs. J Pharm Sci 2016; 105:460-475. [PMID: 26869412 DOI: 10.1016/j.xphs.2015.11.015] [Citation(s) in RCA: 493] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 10/01/2015] [Accepted: 10/30/2015] [Indexed: 01/01/2023]
Abstract
Modification of biopharmaceutical molecules by covalent conjugation of polyethylene glycol (PEG) molecules is known to enhance pharmacologic and pharmaceutical properties of proteins and other large molecules and has been used successfully in 12 approved drugs. Both linear and branched-chain PEG reagents with molecular sizes of up to 40 kDa have been used with a variety of different PEG derivatives with different linker chemistries. This review describes the properties of PEG itself, the history and evolution of PEGylation chemistry, and provides examples of PEGylated drugs with an established medical history. A trend toward the use of complex PEG architectures and larger PEG polymers, but with very pure and well-characterized PEG reagents is described. Nonclinical toxicology findings related to PEG in approved PEGylated biopharmaceuticals are summarized. The effect attributed to the PEG part of the molecules as observed in 5 of the 12 marketed products was cellular vacuolation seen microscopically mainly in phagocytic cells which is likely related to their biological function to absorb and remove particles and macromolecules from blood and tissues. Experience with marketed PEGylated products indicates that adverse effects in toxicology studies are usually related to the active part of the drug but not to the PEG moiety.
Collapse
Affiliation(s)
| | - Mary J Bossard
- Nektar Therapeutics, Department of Polymer Chemistry, Huntsville, Alabama 35801-5914
| | | | - Inge A Ivens
- Bayer, Toxicology, San Francisco, California 94158
| |
Collapse
|
28
|
Steinbach T, Becker G, Spiegel A, Figueiredo T, Russo D, Wurm FR. Reversible Bioconjugation: Biodegradable Poly(phosphate)-Protein Conjugates. Macromol Biosci 2016; 17. [PMID: 27943530 DOI: 10.1002/mabi.201600377] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/09/2016] [Indexed: 11/05/2022]
Abstract
Protein-polymer conjugates are widely used to improve the pharmacokinetic properties of therapeutic proteins. Commercially available conjugates employ poly(ethylene glycol) (PEG) as the protective polymer; however, PEG has a number of shortcomings, including non-biodegradability and immunogenicity, that call for the development of alternatives. Here, the synthesis of biodegradable poly(phosphate), that is, poly(ethyl ethylene phosphate) (PEEP), by organo-catalyzed anionic ring-opening polymerization exhibiting dispersity values Ð < 1.3 is reported. Polymers with molecular weights between 2000 and 33 200 g mol-1 are then ω-functionalized with a succinimidyl carbonate group and subsequently conjugated to model proteins. These are the first conjugates based on polyphosphates which degraded upon exposure to phosphodiesterase. As is the case for PEGylated therapeutics, residual in vitro activity of the PPEylated conjugates depends on the extent of protein modification. These results suggest that PEEP exhibits the desired properties of a biopolymer for use in next generation, fully degradable drug delivery systems.
Collapse
Affiliation(s)
- Tobias Steinbach
- Max-Planck-Institut für Polymerforschung, Ackermannweg 10, 55128, Mainz, Germany.,Graduate School Material Science in Mainz, Staudinger Weg 9, 55128, Mainz, Germany
| | - Greta Becker
- Max-Planck-Institut für Polymerforschung, Ackermannweg 10, 55128, Mainz, Germany.,Graduate School Material Science in Mainz, Staudinger Weg 9, 55128, Mainz, Germany
| | - Alina Spiegel
- Max-Planck-Institut für Polymerforschung, Ackermannweg 10, 55128, Mainz, Germany
| | - Tamiris Figueiredo
- CNR Istituto Officina dei Materiali c/o Institut Laue Langevin, 38000, Grenoble, France
| | - Daniela Russo
- CNR Istituto Officina dei Materiali c/o Institut Laue Langevin, 38000, Grenoble, France.,Institut Lumière Matière, Université de Lyon 1, 69622, Lyon, France
| | - Frederik R Wurm
- Max-Planck-Institut für Polymerforschung, Ackermannweg 10, 55128, Mainz, Germany
| |
Collapse
|
29
|
Collins J, Kempe K, Wilson P, Blindauer CA, McIntosh MP, Davis TP, Whittaker MR, Haddleton DM. Stability Enhancing N-Terminal PEGylation of Oxytocin Exploiting Different Polymer Architectures and Conjugation Approaches. Biomacromolecules 2016; 17:2755-66. [PMID: 27419537 DOI: 10.1021/acs.biomac.6b00919] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Oxytocin, a cyclic nine amino acid neurohypophyseal hormone therapeutic, is effectively used in the control of postpartum hemorrhaging (PPH) and is on the WHO List of Essential Medicines. However, oxytocin has limited shelf life stability in aqueous solutions, particularly at temperatures in excess of 25 °C and injectable aqueous oxytocin formulations require refrigeration (<8 °C). This is particularly problematic in the hot climates often found in many developing countries where daytime temperatures can exceed 40 °C and where reliable cold-chain storage is not always achievable. The purpose of this study was to develop N-terminal amine targeted PEGylation strategies utilizing both linear PEG and polyPEG "comb" polymers as an effective method for stabilizing solution formulations of this peptide for prolonged storage in the absence of efficient cold-chain storage. The conjugation chemistries investigated herein include irreversible amine targeted conjugation methods utilizing NHS ester and aldehyde reductive amination chemistry. Additionally, one reversible conjugation method using a Schiff base approach was explored to allow for the release of the native peptide, thus, ensuring that biological activity remains unaffected. The reversibility of this approach was investigated for the different polymer architectures, alongside a nonpolymer oxytocin analogue to monitor how pH can tune native peptide release. Elevated temperature degradation studies of the polymer conjugates were evaluated to assess the stability of the PEGylated analogues in comparison to the native peptide in aqueous formulations to mimic storage conditions in developing nations and regions where storage under appropriate conditions is challenging.
Collapse
Affiliation(s)
- Jennifer Collins
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom
| | - Kristian Kempe
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Paul Wilson
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Claudia A Blindauer
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom
| | - Michelle P McIntosh
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Michael R Whittaker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - David M Haddleton
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| |
Collapse
|
30
|
Reichert C, Borchard G. Noncovalent PEGylation, An Innovative Subchapter in the Field of Protein Modification. J Pharm Sci 2016; 105:386-390. [PMID: 26523632 DOI: 10.1002/jps.24692] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/03/2015] [Accepted: 09/24/2015] [Indexed: 12/17/2022]
Abstract
Attachment of a chain of poly(ethylene glycol) (PEG) to a therapeutic protein, a process widely known as PEGylation, can lead to several beneficial effects. It has the potential to significantly delay aggregation of the protein by steric shielding, a frequently encountered issue in the development of protein drugs. Moreover, it can modify the pharmacokinetic profile of the PEGylated protein by delaying renal excretion, leading to a longer half-life (t1/2) of the drug. By steric hindrance, it can also inhibit interactions between the protein drug and proteases as well as the host immune system, thereby inhibiting inactivation of the PEGylated protein and also attenuating its immunogenicity. Unfortunately, the effect of steric hindrance also applies to protein drug-target interaction, leading to a (partial) loss of efficacy. In order to avoid this undesirable effect, several efforts have been made to link PEG to a protein in a noncovalent way, providing the protein with several of the beneficial effects of PEGylation while also taking advantage of its native affinity to its target.
Collapse
Affiliation(s)
- Christian Reichert
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Gerrit Borchard
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland.
| |
Collapse
|
31
|
Baines A, Martin P, Rorie C. Current and Emerging Targeting Strategies for Treatment of Pancreatic Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:277-320. [DOI: 10.1016/bs.pmbts.2016.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Delplace V, Nicolas J. Degradable vinyl polymers for biomedical applications. Nat Chem 2015; 7:771-84. [PMID: 26391076 DOI: 10.1038/nchem.2343] [Citation(s) in RCA: 241] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 08/04/2015] [Indexed: 12/23/2022]
Abstract
Vinyl polymers have been the focus of intensive research over the past few decades and are attractive materials owing to their ease of synthesis and their broad diversity of architectures, compositions and functionalities. Their carbon-carbon backbones are extremely resistant to degradation, however, and this property limits their uses. Degradable polymers are an important field of research in polymer science and have been used in a wide range of applications spanning from (nano)medicine to microelectronics and environmental protection. The development of synthetic strategies to enable complete or partial degradation of vinyl polymers is, therefore, of great importance because it will offer new opportunities for the application of these materials. This Review captures the most recent and promising approaches to the design of degradable vinyl polymers and discusses the potential of these materials for biomedical applications.
Collapse
Affiliation(s)
- Vianney Delplace
- Institut Galien Paris-Sud, CNRS UMR 8612, Univ Paris-Sud, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry cedex, France
| | - Julien Nicolas
- Institut Galien Paris-Sud, CNRS UMR 8612, Univ Paris-Sud, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry cedex, France
| |
Collapse
|
33
|
Herzberger J, Niederer K, Pohlit H, Seiwert J, Worm M, Wurm FR, Frey H. Polymerization of Ethylene Oxide, Propylene Oxide, and Other Alkylene Oxides: Synthesis, Novel Polymer Architectures, and Bioconjugation. Chem Rev 2015; 116:2170-243. [PMID: 26713458 DOI: 10.1021/acs.chemrev.5b00441] [Citation(s) in RCA: 478] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The review summarizes current trends and developments in the polymerization of alkylene oxides in the last two decades since 1995, with a particular focus on the most important epoxide monomers ethylene oxide (EO), propylene oxide (PO), and butylene oxide (BO). Classical synthetic pathways, i.e., anionic polymerization, coordination polymerization, and cationic polymerization of epoxides (oxiranes), are briefly reviewed. The main focus of the review lies on more recent and in some cases metal-free methods for epoxide polymerization, i.e., the activated monomer strategy, the use of organocatalysts, such as N-heterocyclic carbenes (NHCs) and N-heterocyclic olefins (NHOs) as well as phosphazene bases. In addition, the commercially relevant double-metal cyanide (DMC) catalyst systems are discussed. Besides the synthetic progress, new types of multifunctional linear PEG (mf-PEG) and PPO structures accessible by copolymerization of EO or PO with functional epoxide comonomers are presented as well as complex branched, hyperbranched, and dendrimer like polyethers. Amphiphilic block copolymers based on PEO and PPO (Poloxamers and Pluronics) and advances in the area of PEGylation as the most important bioconjugation strategy are also summarized. With the ever growing toolbox for epoxide polymerization, a "polyether universe" may be envisaged that in its structural diversity parallels the immense variety of structural options available for polymers based on vinyl monomers with a purely carbon-based backbone.
Collapse
Affiliation(s)
- Jana Herzberger
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany
| | - Kerstin Niederer
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Hannah Pohlit
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany.,Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany.,Department of Dermatology, University Medical Center , Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Jan Seiwert
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Matthias Worm
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany
| | - Frederik R Wurm
- Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany.,Max Planck Institute for Polymer Research , Ackermannweg 10, D-55128 Mainz, Germany
| | - Holger Frey
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany
| |
Collapse
|
34
|
Liu J, Postupalenko V, Duskey JT, Palivan CG, Meier W. pH-Triggered Reversible Multiple Protein-Polymer Conjugation Based on Molecular Recognition. J Phys Chem B 2015; 119:12066-73. [PMID: 26291123 DOI: 10.1021/acs.jpcb.5b06637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Polymer conjugation for protein-based therapeutics has been developed extensively, but it still suffers from conjugation leading to decrease in protein activity and generates complexes with limited diversity due to general classical systems only incorporating one protein per each complex. Here we introduce a site-specific noncovalent protein-polymer conjugation, which can reduce the heterogeneity of the conjugates without disrupting protein function, while allowing for the modulation of binding affinity and stability, affecting the pH dependent binding of the number of proteins per polymer. We compared classical one protein-polymer conjugates with multiple protein-polymer conjugates using His-tagged enhanced yellow fluorescence protein (His6-eYFP) and metal-coordinated tris-nitrilotriacetic acid (trisNTA-Me(n+)) in a site-specific way. trisNTA-Me(n+)-His6 acts as a reversible linker with pH-triggered release of functional protein from the trisNTA-functionalized copolymers. The nature of the selected Me(n+) and number of available trisNTA-Me(n+) on poly(N-isopropylacrylamide-co-tris-nitrilotriacetic acid acrylamide) (PNTn) copolymers enables predictable modulation of the conjugates binding affinity (0.09-1.35 μM), stability, cell toxicity, and pH responsiveness. This represents a promising platform that allows direct control over the properties of multiple protein-polymer conjugates compared to the classical single protein-polymer conjugates.
Collapse
Affiliation(s)
- Juan Liu
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, Basel 4056, Switzerland
| | - Viktoriia Postupalenko
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, Basel 4056, Switzerland
| | - Jason T Duskey
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, Basel 4056, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, Basel 4056, Switzerland
| | - Wolfgang Meier
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, Basel 4056, Switzerland
| |
Collapse
|
35
|
Li Z, Han X, Zhai Y, Lian H, Zhang D, Zhang W, Wang Y, He Z, Liu Z, Sun J. Critical determinant of intestinal permeability and oral bioavailability of pegylated all trans -retinoic acid prodrug-based nanomicelles: Chain length of poly (ethylene glycol) corona. Colloids Surf B Biointerfaces 2015; 130:133-40. [DOI: 10.1016/j.colsurfb.2015.03.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/15/2015] [Accepted: 03/16/2015] [Indexed: 11/25/2022]
|
36
|
Skurikhin EG, Pershina OV, Reztsova AM, Ermakova NN, Khmelevskaya ES, Krupin VA, Stepanova IE, Artamonov AV, Bekarev AA, Madonov PG, Dygai AM. Modulation of bleomycin-induced lung fibrosis by pegylated hyaluronidase and dopamine receptor antagonist in mice. PLoS One 2015; 10:e0125065. [PMID: 25927611 PMCID: PMC4415936 DOI: 10.1371/journal.pone.0125065] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 03/20/2015] [Indexed: 12/30/2022] Open
Abstract
Hyaluronidases are groups of enzymes that degrade hyaluronic acid (HA). To stop enzymatic hydrolysis we modified testicular hyaluronidase (HYAL) by activated polyethylene oxide with the help of electron-beam synthesis. As a result we received pegylated hyaluronidase (pegHYAL). Spiperone is a selective D2 dopamine receptor antagonist. It was demonstrated on the model of a single bleomycin damage of alveolar epithelium that during the inflammatory phase monotherapy by pegHYAL or spiperone reduced the populations of hematopoietic stem /progenitor cells in the lung parenchyma. PegHYAL also reduced the levels of transforming growth factor (TGF)-β, interleukin (IL)-1β, tumor necrosis factor (TNF)-α in the serum and lungs, while spiperone reduced the level of the serum IL-1β. Polytherapy by spiperone and pegHYAL caused the increase of the quantity of hematopoietic stem/ progenitor cells in the lungs. Such an influx of blood cell precursors was observed on the background of considerable fall level of TGF-β and the increase level of TNF-α in the serum and lungs. These results show pegHYAL reduced the bleomycin-induced fibrosis reaction (production and accumulation of collagen) in the lung parenchyma. This effect was observed at a single and repetitive bleomycin damage of alveolar epithelium, the antifibrotic activity of pegHYAL surpassing the activity of testicular HYAL. The antifibrotic effect of pegHYAL is enhanced by an additional instillation of spiperone. Therapy by pegHYAL causes the flow of CD31‒ CD34‒ CD45‒ CD44+ CD73+ CD90+ CD106+-cells into the fibrous lungs. These cells are incapable of differentiating into fibroblast cells. Spiperone instillation separately or together with pegHYAL reduced the MSC-like cells considerably. These data enable us to assume, that pegHYAL is a new and promising instrument both for preventive and therapy of toxic pneumofibrosis. The blockage of D2 dopamine receptors with the following change of hyaluronan matrix can be considered as a new strategy in treatment of pneumofibrosis.
Collapse
Affiliation(s)
- Evgenii Germanovich Skurikhin
- Department of Pathophysiology and Regenerative Medicine, Research Institute of Pharmacology and Regenerative Medicine named after E.D. Goldberg, Tomsk, Russia
| | - Olga Victorovna Pershina
- Department of Pathophysiology and Regenerative Medicine, Research Institute of Pharmacology and Regenerative Medicine named after E.D. Goldberg, Tomsk, Russia
- * E-mail:
| | - Alena Mikhaylovna Reztsova
- Department of Pathophysiology and Regenerative Medicine, Research Institute of Pharmacology and Regenerative Medicine named after E.D. Goldberg, Tomsk, Russia
| | - Natalia Nikolaevna Ermakova
- Department of Pathophysiology and Regenerative Medicine, Research Institute of Pharmacology and Regenerative Medicine named after E.D. Goldberg, Tomsk, Russia
| | - Ekaterina Sergeevna Khmelevskaya
- Department of Pathophysiology and Regenerative Medicine, Research Institute of Pharmacology and Regenerative Medicine named after E.D. Goldberg, Tomsk, Russia
| | - Vycheslav Andreevich Krupin
- Department of Pathophysiology and Regenerative Medicine, Research Institute of Pharmacology and Regenerative Medicine named after E.D. Goldberg, Tomsk, Russia
| | - Inna Ernestovna Stepanova
- Department of Pathophysiology and Regenerative Medicine, Research Institute of Pharmacology and Regenerative Medicine named after E.D. Goldberg, Tomsk, Russia
| | | | | | | | - Alexander Mikhaylovich Dygai
- Department of Pathophysiology and Regenerative Medicine, Research Institute of Pharmacology and Regenerative Medicine named after E.D. Goldberg, Tomsk, Russia
| |
Collapse
|
37
|
Collins J, Tanaka J, Wilson P, Kempe K, Davis TP, McIntosh MP, Whittaker MR, Haddleton DM. In Situ Conjugation of Dithiophenol Maleimide Polymers and Oxytocin for Stable and Reversible Polymer–Peptide Conjugates. Bioconjug Chem 2015; 26:633-8. [DOI: 10.1021/bc5006202] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Jennifer Collins
- Department
of Chemistry, University of Warwick, CV4 7AL, Coventry, United Kingdom
| | - Joji Tanaka
- Department
of Chemistry, University of Warwick, CV4 7AL, Coventry, United Kingdom
| | - Paul Wilson
- Department
of Chemistry, University of Warwick, CV4 7AL, Coventry, United Kingdom
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kristian Kempe
- Department
of Chemistry, University of Warwick, CV4 7AL, Coventry, United Kingdom
| | - Thomas P. Davis
- Department
of Chemistry, University of Warwick, CV4 7AL, Coventry, United Kingdom
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Michelle P. McIntosh
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Michael R. Whittaker
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - David M. Haddleton
- Department
of Chemistry, University of Warwick, CV4 7AL, Coventry, United Kingdom
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
38
|
Gong Y, Leroux JC, Gauthier MA. Releasable Conjugation of Polymers to Proteins. Bioconjug Chem 2015; 26:1172-81. [DOI: 10.1021/bc500611k] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yuhui Gong
- Swiss
Federal Institute of Technology Zurich (ETHZ), Department of Chemistry
and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg
1−5/10, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Swiss
Federal Institute of Technology Zurich (ETHZ), Department of Chemistry
and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg
1−5/10, 8093 Zurich, Switzerland
| | - Marc A. Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Centre, 1650 boul. Lionel-Boulet, Varennes, J3X 1S2, Canada
| |
Collapse
|
39
|
Wu Y, Ng DYW, Kuan SL, Weil T. Protein–polymer therapeutics: a macromolecular perspective. Biomater Sci 2015. [DOI: 10.1039/c4bm00270a] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The development of protein–polymer hybrids emerged several decades ago with the vision that their synergistic combination will offer macromolecular hybrids with manifold features to succeed as the next generation therapeutics.
Collapse
Affiliation(s)
- Yuzhou Wu
- Institute of Organic Chemistry III
- Macromolecular Chemistry
- Albert-Einstein-Allee 11
- 89081 Ulm
- Germany
| | - David Y. W. Ng
- Institute of Organic Chemistry III
- Macromolecular Chemistry
- Albert-Einstein-Allee 11
- 89081 Ulm
- Germany
| | - Seah Ling Kuan
- Institute of Organic Chemistry III
- Macromolecular Chemistry
- Albert-Einstein-Allee 11
- 89081 Ulm
- Germany
| | - Tanja Weil
- Institute of Organic Chemistry III
- Macromolecular Chemistry
- Albert-Einstein-Allee 11
- 89081 Ulm
- Germany
| |
Collapse
|
40
|
Drapala PW, Jiang B, Chiu YC, Mieler WF, Brey EM, Kang-Mieler JJ, Pérez-Luna VH. The effect of glutathione as chain transfer agent in PNIPAAm-based thermo-responsive hydrogels for controlled release of proteins. Pharm Res 2014; 31:742-53. [PMID: 24022682 DOI: 10.1007/s11095-013-1195-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/21/2013] [Indexed: 12/29/2022]
Abstract
PURPOSE To control degradation and protein release using thermo-responsive hydrogels for localized delivery of anti-angiogenic proteins. METHODS Thermo-responsive hydrogels derived from N-isopropylacrylamide (NIPAAm) and crosslinked with poly(ethylene glycol)-co-(L-lactic acid) diacrylate (Acry-PLLA-b-PEG-b-PLLA-Acry) were synthesized via free radical polymerization in the presence of glutathione, a chain transfer agent (CTA) added to modulate their degradation and release properties. Immunoglobulin G (IgG) and the recombinant proteins Avastin® and Lucentis® were encapsulated in these hydrogels and their release was studied. RESULTS The encapsulation efficiency of IgG was high (75-87%) and decreased with CTA concentration. The transition temperature of these hydrogels was below physiological temperature, which is important for minimally invasive therapies involving these materials. The toxicity from unreacted monomers and free radical initiators was eliminated with a minimum of three buffer extractions. Addition of CTA accelerated degradation and resulted in complete protein release. Glutathione caused the degradation products to become solubilized even at 37°C. Hydrogels prepared without glutathione did not disintegrate nor released protein completely after 3 weeks at 37°C. PEGylation of IgG postponed the burst release effect. Avastin® and Lucentis® released from degraded hydrogels retained their biological activity. CONCLUSIONS These systems offer a promising platform for the localized delivery of proteins.
Collapse
|
41
|
PEG — A versatile conjugating ligand for drugs and drug delivery systems. J Control Release 2014; 192:67-81. [DOI: 10.1016/j.jconrel.2014.06.046] [Citation(s) in RCA: 471] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/23/2014] [Accepted: 06/24/2014] [Indexed: 01/07/2023]
|
42
|
van Dongen M, Dougherty CA, Banaszak Holl MM. Multivalent polymers for drug delivery and imaging: the challenges of conjugation. Biomacromolecules 2014; 15:3215-34. [PMID: 25120091 PMCID: PMC4157765 DOI: 10.1021/bm500921q] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/06/2014] [Indexed: 12/11/2022]
Abstract
Multivalent polymers offer a powerful opportunity to develop theranostic materials on the size scale of proteins that can provide targeting, imaging, and therapeutic functionality. Achieving this goal requires the presence of multiple targeting molecules, dyes, and/or drugs on the polymer scaffold. This critical review examines the synthetic, analytical, and functional challenges associated with the heterogeneity introduced by conjugation reactions as well as polymer scaffold design. First, approaches to making multivalent polymer conjugations are discussed followed by an analysis of materials that have shown particular promise biologically. Challenges in characterizing the mixed ligand distributions and the impact of these distributions on biological applications are then discussed. Where possible, molecular-level interpretations are provided for the structures that give rise to the functional ligand and molecular weight distributions present in the polymer scaffolds. Lastly, recent strategies employed for overcoming or minimizing the presence of ligand distributions are discussed. This review focuses on multivalent polymer scaffolds where average stoichiometry and/or the distribution of products have been characterized by at least one experimental technique. Key illustrative examples are provided for scaffolds that have been carried forward to in vitro and in vivo testing with significant biological results.
Collapse
Affiliation(s)
- Mallory
A. van Dongen
- Chemistry Department, University of Michigan, Ann Arbor, Michigan 48103, United States
| | - Casey A. Dougherty
- Chemistry Department, University of Michigan, Ann Arbor, Michigan 48103, United States
| | - Mark M. Banaszak Holl
- Chemistry Department, University of Michigan, Ann Arbor, Michigan 48103, United States
| |
Collapse
|
43
|
Ye WL, Du JB, Zhang BL, Na R, Song YF, Mei QB, Zhao MG, Zhou SY. Cellular uptake and antitumor activity of DOX-hyd-PEG-FA nanoparticles. PLoS One 2014; 9:e97358. [PMID: 24828815 PMCID: PMC4020841 DOI: 10.1371/journal.pone.0097358] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 04/17/2014] [Indexed: 11/24/2022] Open
Abstract
A PEG-based, folate mediated, active tumor targeting drug delivery system using DOX-hyd-PEG-FA nanoparticles (NPs) were prepared. DOX-hyd-PEG-FA NPs showed a significantly faster DOX release in pH 5.0 medium than in pH 7.4 medium. Compared with DOX-hyd-PEG NPs, DOX-hyd-PEG-FA NPs increased the intracellular accumulation of DOX and showed a DOX translocation from lysosomes to nucleus. The cytotoxicity of DOX-hyd-PEG-FA NPs on KB cells was much higher than that of free DOX, DOX-ami-PEG-FA NPs and DOX-hyd-PEG NPs. The cytotoxicity of DOX-hyd-PEG-FA NPs on KB cells was attenuated in the presence of exogenous folic acid. The IC50 of DOX-hyd-PEG-FA NPs and DOX-hyd-PEG NPs on A549 cells showed no significant difference. After DOX-hyd-PEG-FA NPs were intravenously administered, the amount of DOX distributed in tumor tissue was significantly increased, while the amount of DOX distributed in heart was greatly decreased as compared with free DOX. Compared with free DOX, NPs yielded improved survival rate, prolonged life span, delayed tumor growth and reduced the cardiotoxicity in tumor bearing mice model. These results indicated that the acid sensitivity, passive and active tumor targeting abilities were likely to act synergistically to enhance the drug delivery efficiency of DOX-hyd-PEG-FA NPs. Therefore, DOX-hyd-PEG-FA NPs are a promising drug delivery system for targeted cancer therapy.
Collapse
Affiliation(s)
- Wei-liang Ye
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Jiang-bo Du
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Bang-le Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ren Na
- West Changle Sanatorium for Xi'an Army Retired Cadres of Fourth Military Medical University, Xi'an, China
| | - Yan-feng Song
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Qi-bing Mei
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ming-gao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Si-yuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, China
- * E-mail:
| |
Collapse
|
44
|
Zacchigna M, Cateni F, Drioli S, Procida G, Altieri T. PEG-Ursolic Acid Conjugate: Synthesis and In Vitro Release Studies. Sci Pharm 2014; 82:411-21. [PMID: 24959409 PMCID: PMC4065131 DOI: 10.3797/scipharm.1309-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 04/06/2014] [Indexed: 11/22/2022] Open
Abstract
A highly water-soluble macromolecular compound of ursolic acid with monomethoxypoly(ethylene glycol) (mPEG) was prepared. The physicochemical properties and stabilities under different conditions were investigated. By PEG conjugation, greatly increased water solubility was obtained, and the results showed that this conjugate was a potential prodrug for the oral delivery of ursolic acid.
Collapse
Affiliation(s)
- Marina Zacchigna
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy
| | - Francesca Cateni
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy
| | - Sara Drioli
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy
| | - Giuseppe Procida
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy
| | - Tiziano Altieri
- School of Advanced Studies "G. D'Annunzio", University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy
| |
Collapse
|
45
|
Cleavable carbamate linkers for controlled protein delivery from hydrogels. J Control Release 2014; 183:67-76. [PMID: 24680687 DOI: 10.1016/j.jconrel.2014.03.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 11/21/2022]
Abstract
The reversible attachment of proteins to polymers is one potential strategy to control protein release from hydrogels. In this study, we report the reversible attachment of lysozyme to poly(ethylene glycol) (PEG) by degradable carbamate linkers. Phenyl groups with different substituents were used to control the rate of carbamate hydrolysis and the resulting protein release. Sodium dodecyl sulfate polyacrylamide gel electrophoresis showed modification with 1-3 PEG chains per lysozyme molecule. Protein PEGylation and PEG chain elimination occurred without changes in secondary protein structure, as demonstrated by circular dichroism spectroscopy. The lytic activity of lysozyme was restored to 73.4±1.7%-92.5±1.2% during PEG chain elimination. Attached PEG chains were eliminated within 24h to 28days, depending on the used linker molecule. When formulated into hydrogels, a maximum of about 60% of the initial dose was released within 7days to 21days. Linker elimination occurs 'traceless', so that the protein is released in its native, unmodified form. Altogether, we believe that tethering proteins by degradable carbamate linkers is a promising strategy to control their release from hydrogels.
Collapse
|
46
|
Santi DV, Schneider EL, Ashley GW. Macromolecular prodrug that provides the irinotecan (CPT-11) active-metabolite SN-38 with ultralong half-life, low C(max), and low glucuronide formation. J Med Chem 2014; 57:2303-14. [PMID: 24494988 DOI: 10.1021/jm401644v] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We have recently reported a chemical approach for half-life extension that utilizes β-eliminative linkers to attach amine-containing drugs or prodrugs to macromolecules. The linkers release free drug or prodrug over periods ranging from a few hours to over 1 year. We adapted these linkers for use with phenol-containing drugs. Here, we prepared PEG conjugates of the irinotecan (CPT-11) active metabolite SN-38 via a phenyl ether that release the drug with predictable long half-lives. Pharmacokinetic studies in the rat indicate that, in contrast to other SN-38 prodrugs, the slowly released SN-38 shows a very low C(max), is kept above target concentrations for extended periods, and forms very little SN-38 glucuronide (the precursor of enterotoxic SN-38). The low SN-38 glucuronide is attributed to low hepatic uptake of SN-38. These macromolecular prodrugs have unique pharmacokinetic profiles that may translate to less intestinal toxicity and interpatient variability than the SN-38 prodrugs thus far studied.
Collapse
Affiliation(s)
- Daniel V Santi
- ProLynx , 455 Mission Bay Boulevard South, Suite 145, San Francisco, California 94158, United States
| | | | | |
Collapse
|
47
|
Amedei A, Niccolai E, Prisco D. Pancreatic cancer: role of the immune system in cancer progression and vaccine-based immunotherapy. Hum Vaccin Immunother 2014; 10:3354-3368. [PMID: 25483688 PMCID: PMC4514060 DOI: 10.4161/hv.34392] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/25/2014] [Accepted: 08/08/2014] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer (PC) is the 5th leading cause of cancer related death in the developed world with more than 260,000 deaths annually worldwide and with a dismal 5-year survival. Surgery is the only potential hope of cure for PC, but, unfortunately, only 20% PC patients is resectable at the time of diagnosis. Therapeutic research efforts have mainly focused on improvements in radio/ chemo treatments and to date, there are only a few chemotherapeutic agents that have shown to be effective against PC, including gemcitabine with or without abraxane as well as a combination of 5-FU, leucovorin, oxaliplatin and irinotecan (the so-called FOLFIRINOX regimen). The survival of patients treated with these regimens is marginal and hence we are in urgent need of novel therapeutic approaches to treat pancreatic cancer. The success of immunotherapeutic strategies in other cancers and various evidences that pancreatic adenocarcinoma elicits antitumor immune responses, suggest that immunotherapies can be a promising alternative treatment modality for this deadly disease. PC immunotherapy treatments include passive immunotherapeutic approaches, such as the use of effector cells generated in vitro, and active immunotherapeutic strategies, which goal is to stimulate an antitumor response in vivo, by means of vaccination. In this review, we describe the immune suppressive mechanisms of pancreatic cancer and discuss recent preclinical and clinical efforts toward PC immunotherapy, including passive approaches, such as the use of antibodies and active strategies (vaccination), with a special mention of most recent treatment with CRS-207 and GVAX.
Collapse
Key Words
- APC, Antigen Presenting Cells
- CEA, carcinoembryonic antigen
- CTL, Cytotoxic CD8 T cells
- DCs, Dendritic Cells
- ENO1, a-Enolasi
- IDO, Indoleamine 2,3-dioxygenase
- MUC1, Mucin-1
- NK, Natural Killer
- PC, pancreatic cancer
- Th, T helper
- Tregs, Regulatory T cells
- clinical trials
- immune response
- immunotherapy
- mAbs, monoclonal antibodies
- pancreatic cancer
- vaccine
Collapse
Affiliation(s)
- Amedeo Amedei
- Department of Experimental and Clinical Internal Medicine; University of Florence; Florence, Italy
- Department of Biomedicine; Azienda Ospedaliera Universitaria Careggi (AOUC); Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Internal Medicine; University of Florence; Florence, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Internal Medicine; University of Florence; Florence, Italy
- Department of Biomedicine; Azienda Ospedaliera Universitaria Careggi (AOUC); Florence, Italy
| |
Collapse
|
48
|
Xia Y, Wang X, Wang Y, Wang Y. Full pH-range responsive hyperbranched polyethers: synthesis and responsiveness. Polym Chem 2014. [DOI: 10.1039/c4py00284a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In order to impart full pH-range responsiveness within biocompatible hyperbranched polyethers, new amphiphilic polyethers, i.e. HPMHO–Amines and HPMHO–Carboxys, which have a molecular structure similar to hyperbranched PEG, were prepared through ring-opening polymerization and modified by amination or carboxylation.
Collapse
Affiliation(s)
- Yumin Xia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Materials Science and Engineering
- Donghua University
- Shanghai, People's Republic of China
| | - Xinhang Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Materials Science and Engineering
- Donghua University
- Shanghai, People's Republic of China
| | - Yanping Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Materials Science and Engineering
- Donghua University
- Shanghai, People's Republic of China
| | - Yimin Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Materials Science and Engineering
- Donghua University
- Shanghai, People's Republic of China
| |
Collapse
|
49
|
Ikeda Y, Nagasaki Y. Impacts of PEGylation on the gene and oligonucleotide delivery system. J Appl Polym Sci 2013. [DOI: 10.1002/app.40293] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Yutaka Ikeda
- Department of Materials Sciences; Graduate School of Pure and Applied Sciences, University of Tsukuba; Ibaraki 305-8573 Japan
| | - Yukio Nagasaki
- Department of Materials Sciences; Graduate School of Pure and Applied Sciences, University of Tsukuba; Ibaraki 305-8573 Japan
- Master's School of Medical Sciences; Graduate School of Comprehensive Human Sciences, University of Tsukuba; Ibaraki 305-8573 Japan
- Satellite Laboratory; International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute of Materials Science (NIMS); Tennodai 1-1-1, Tsukuba Ibaraki 305-8573 Japan
| |
Collapse
|
50
|
Soshee A, Zürcher S, Spencer ND, Halperin A, Nizak C. General in vitro method to analyze the interactions of synthetic polymers with human antibody repertoires. Biomacromolecules 2013; 15:113-21. [PMID: 24328191 DOI: 10.1021/bm401360y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent reports on the hitherto underestimated antigenicity of poly(ethylene glycol) (PEG), which is widely used for pharmaceutical applications, highlight the need for efficient testing of polymer antigenicity and for a better understanding of its molecular origins. With this goal in mind, we have used the phage-display technique to screen large, recombinant antibody repertoires of human origin in vitro for antibodies that bind poly(vinylpyrrolidone) (PVP). PVP is a neutral synthetic polymer of industrial and clinical interest that is also a well-known model antigen in animal studies, thus allowing the comparison of in vitro and in vivo responses. We have identified 44 distinct antibodies that bind specifically to PVP. Competitive binding assays show that the PVP-antibody binding constant is proportional to the polymerization degree of PVP and that specific binding is detected down to the vinylpyrrolidone (VP) monomer level. Statistical analysis of anti-PVP antibody sequences identifies an amino-acid motif that is shared by many phage-display-selected anti-PVP antibodies that are similar to a previously described natural anti-PVP antibody. This suggests a role for this motif in specific antibody/PVP interactions. Interestingly, sequence analysis also suggests that only a single antibody chain containing this shared motif is responsible for antibody binding to PVP, as confirmed upon systematic deletion of either antibody chain for 90% of selected anti-PVP antibodies. Overall, a large number of antibodies in the human repertoires we have screened bind specifically to PVP through a small number of shared amino acid motifs, and preliminary comparison points to significant correlations between the sequences of phage-display-selected anti-PVP antibodies and their natural counterparts isolated from immunized mice in previous studies. This study pioneers the use of antibody phage-display to explore the antigenicity of biotechnologically relevant polymers. It also paves the way for a fast, cost-effective, and systematic in vitro analysis, thus reducing the need for animal immunization experiments. Moreover, identifying the encoding DNA sequence of polymer-binding antibodies via phage-display enables future applications of a molecular biology approach to protein-polymer conjugation, based on protein-antibody fusion.
Collapse
Affiliation(s)
- Anandakumar Soshee
- Laboratory of Interdisciplinary Physics, UMR5588 Grenoble Université 1/CNRS , Grenoble, France
| | | | | | | | | |
Collapse
|