1
|
Yaseen Z, Nandave M, Sharma L. Anti-diabetic Biologicals: Exploring the Role of Different Analytical Techniques. Crit Rev Anal Chem 2025:1-22. [PMID: 40088445 DOI: 10.1080/10408347.2025.2472793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Antidiabetic biologicals (ADBs) have revolutionized the treatment of diabetes mellitus, once considered incurable through conventional medicine. These biological products, derived from natural sources via extraction, semi-synthesis, or recombinant DNA technology, include insulin and its analogs, GLP-1 receptor agonists, amylin analogs, and the recently approved monoclonal antibody teplizumab. Regulatory authorities worldwide have established QC parameters outlined in pharmacopoeias, alongside analytical techniques to ensure their safety and efficacy. This review focuses on the analytical techniques used to assess QC parameters of ADBs, including chromatographic methods, spectroscopic techniques, capillary electrophoresis, immunoassays, and endotoxin testing. Key parameters such as identification, potency, purity, and impurity profiling are thoroughly examined. The paper provides a comprehensive and up-to-date compilation of QC requirements and methodologies, along with a detailed comparison of analytical techniques. In doing so, it highlights their advantages and limitations, offering valuable insights for researchers and regulatory professionals involved in selecting suitable methods for QC assessment and understanding the complexities of ADBs evaluation. Furthermore, the article discusses the paramount importance of QC and future perspectives, emphasizing the transition to advanced versions of current techniques driven by the need for efficiency and reliability in quality testing.
Collapse
Affiliation(s)
- Zahid Yaseen
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
- Department of Pharmaceutical Biotechnology, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Mukesh Nandave
- Department of Pharmacology, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| |
Collapse
|
2
|
Armstrong L, Chang SL, Clements N, Hirani Z, Kimberly LB, Odoi-Adams K, Suating P, Taylor HF, Trauth SA, Urbach AR. Molecular recognition of peptides and proteins by cucurbit[ n]urils: systems and applications. Chem Soc Rev 2024; 53:11519-11556. [PMID: 39415690 PMCID: PMC11484504 DOI: 10.1039/d4cs00569d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Indexed: 10/19/2024]
Abstract
The development of methodology for attaching ligand binding sites to proteins of interest has accelerated biomedical science. Such protein tags have widespread applications as well as properties that significantly limit their utility. This review describes the mechanisms and applications of supramolecular systems comprising the synthetic receptors cucurbit[7]uril (Q7) or cucurbit[8]uril (Q8) and their polypeptide ligands. Molecular recognition of peptides and proteins occurs at sites of 1-3 amino acids with high selectivity and affinity via several distinct mechanisms, which are supported by extensive thermodynamic and structural studies in aqueous media. The commercial availability, low cost, high stability, and biocompatibility of these synthetic receptors has led to the development of myriad applications. This comprehensive review compiles the molecular recognition studies and the resulting applications with the goals of providing a valuable resource to the community and inspiring the next generation of innovation.
Collapse
Affiliation(s)
- Lilyanna Armstrong
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| | - Sarah L Chang
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| | - Nia Clements
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| | - Zoheb Hirani
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| | - Lauren B Kimberly
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| | - Keturah Odoi-Adams
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK, 73096, USA
| | - Paolo Suating
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| | - Hailey F Taylor
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| | - Sara A Trauth
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| | - Adam R Urbach
- Department of Chemistry, Trinity University, San Antonio, TX, 78212, USA.
| |
Collapse
|
3
|
Zhang Y, Austin MJ, Chou DHC. Insulin Stabilization Designs for Enhanced Therapeutic Efficacy and Accessibility. Acc Chem Res 2024; 57:3303-3315. [PMID: 39466175 DOI: 10.1021/acs.accounts.4c00500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Insulin has remained indispensable in the treatment of diabetes since it was first discovered in 1921. Unlike small molecular drugs, insulin and other protein drugs are prone to degradation when exposed to elevated temperatures, mechanical agitation during transportation, and prolonged storage periods. Therefore, strict cold-chain management is crucial for the insulin supply, requiring significant resources, which can limit the access to insulin, particularly in low-income areas. Moreover, although insulin formulations have advanced tremendously in the last century, insulin treatment still imposes a challenging regimen and provides suboptimal outcomes for the majority of patients. There is an increasing focus on pursuing improved pharmacology, specifically on safer, more user-friendly insulin therapies that minimize the self-management burden. These challenges underscore the need for developing novel insulin formulations with improved stability that are compatible with advanced insulin therapy. Insulin stabilization can be achieved through either chemical modification of insulin or formulation component design. Inspired by insulin-like peptides from invertebrates, we have developed novel stable insulin analogs based on a fundamental understanding of the insulin receptor engagement for insulin bioactivity. We created a novel four-disulfide insulin analog with high aggregation stability and potency by introducing a fourth disulfide bond between a C-terminal extended insulin A-chain and residues near the C-terminus of the B-chain. In an effort to stabilize insulin in its monomeric state to develop ultrafast-acting insulin with rapid absorption upon injection, we have developed a series of structurally miniaturized yet fully active insulin analogs that do not form dimers due to the lack of the canonical B-chain C-terminal octapeptide. Additionally, our study provided strategies for expanding the scope of cucurbit[7]uril (CB[7])-assisted insulin stabilization by engineering safe and biodegradable CB[7]-zwitterionic polypeptide excipients. We also explored insulin N-terminal substitution methods to achieve pH-dependent insulin stabilization without prolonging the duration of action. This Account describes our exploration of engineering stable insulin analogs and formulation design strategies for stabilizing insulin in aqueous solutions. Beyond conventional stabilization strategies for insulin injections, the unmet challenges and recent innovations in insulin stabilization are discussed, addressing the growing demand for alternative, less invasive routes of insulin administration. Additionally, we aim to provide a thorough overview of insulin stabilization from the perspective of commercially available insulin drugs and common pharmaceutical engineering practices in the industry. We also highlight unresolved insulin stabilization challenges and ongoing research strategies. We anticipate that further emphasis on collective efforts of protein engineering, pharmaceutical formulation design, and drug delivery will inform the development of stable and advanced insulin therapy.
Collapse
Affiliation(s)
- Yanxian Zhang
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Maxwell Jack Austin
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Danny Hung-Chieh Chou
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
4
|
Foster L, Foppiani JA, Xun H, Lee D, Utz B, Hernandez Alvarez A, Domingo-Escobar MJ, Taritsa IC, Gavlasova D, Lee TC, Lin GJ, Choudry U, Lin SJ. Zinc in Dermatology: Exploring Its Emerging Role in Enhancing Botulinum Toxin Formulations and Clinical Efficacy. Curr Issues Mol Biol 2024; 46:12088-12098. [PMID: 39590311 PMCID: PMC11593192 DOI: 10.3390/cimb46110717] [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: 08/14/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
This literature review provides a novel exploration of zinc's multifaceted roles in dermatology, with a particular focus on its potential integration into botulinum toxin formulations-an area that remains relatively underexplored in clinical practice. Zinc is widely recognized for its critical functions in skin health, including morphogenesis, regeneration, and protection, and its use in aesthetic medicine offers a unique opportunity for innovation. Specifically, incorporating zinc into botulinum toxin formulations could enhance the efficacy and stability of these treatments. Although zinc has historically been used in topical dermatological products and systemic health interventions, its potential in cosmetic preparations, such as anti-aging therapies or non-invasive aesthetic treatments, remains under-researched. Emerging patents suggest promising formulations combining zinc with botulinum toxin that may improve product stability and extend therapeutic effects. While current studies on oral zinc supplementation present mixed results concerning its ability to prolong botulinum toxin effects, this underscores the need for more rigorous investigation in the realm of aesthetic medicine. Zinc's well-established role in stabilizing dermatological products, such as sunscreens, and its applications in wound healing and skin regeneration, further highlights its potential for broader therapeutic uses beyond cosmetic applications. This review identifies a critical gap in the literature and calls for future research to optimize zinc concentrations and delivery methods specifically for aesthetic medical procedures, offering new insights into improving dermatological treatments beyond the scope of traditional cosmetic preparations.
Collapse
Affiliation(s)
- Lacey Foster
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jose A. Foppiani
- Department of Plastic and Reconstructive Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Helen Xun
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Daniela Lee
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Begum Utz
- Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey
| | - Angelica Hernandez Alvarez
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Maria J. Domingo-Escobar
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Iulianna C. Taritsa
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Dominika Gavlasova
- Institute of Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | | | - Gavin J. Lin
- Nobles and Greenough School, Dedham, MA 02026, USA
| | - Umar Choudry
- Department of Plastic and Reconstructive Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Samuel J. Lin
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Webber MJ. Engineering a Pathway to Glucose-Responsive Therapeutics. Diabetes 2024; 73:1032-1038. [PMID: 38608241 DOI: 10.2337/dbi23-0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024]
Abstract
In 2014, the American Diabetes Association instituted a novel funding paradigm to support diabetes research through its Pathway to Stop Diabetes program. This program took a multifaceted approach to providing key funding to diabetes researchers to advance a broad spectrum of research programs on all aspects of understanding, managing, and treating diabetes. Here, the personal perspective of a 2019 Pathway Accelerator awardee is offered, describing a research program seeking to advance a materials-centered approach to engineering glucose-responsive devices and new delivery tools for better therapeutic outcomes in treating diabetes. This is offered alongside a personal reflection on 5 years of support from the ADA Pathway Program. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN
| |
Collapse
|
6
|
Wang YC, Bai SC, Ye WL, Jiang J, Li G. Recent Progress in Site-Selective Modification of Peptides and Proteins Using Macrocycles. Bioconjug Chem 2024; 35:277-285. [PMID: 38417023 DOI: 10.1021/acs.bioconjchem.3c00534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
Peptides and proteins undergo crucial modifications to alter their physicochemical properties to expand their applications in diverse fields. Various techniques, such as unnatural amino acid incorporation, enzyme catalysis, and chemoselective methods, have been employed for site-selective peptide and protein modification. While traditional methods remain valuable, advancement in host-guest chemistry introduces innovative and promising approaches for the selective modification of peptides and proteins. Macrocycles exhibit robust binding affinities, particularly with natural amino acids, which facilitates their use in selectively binding to specific sequences. This distinctive property endows macrocycles with the potential for modification of target peptides and proteins. This review provides a comprehensive overview of strategies utilizing macrocycles for the selective modification of peptides and proteins. These strategies unlock new possibilities for constructing antibody-drug conjugates and stabilizing volatile medications.
Collapse
Affiliation(s)
- Ye-Cheng Wang
- Fuzhou Institute of Oceanography, College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian 350108, China
- College of Chemical Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Si-Cong Bai
- School of Future Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Wei-Lin Ye
- Fuzhou Institute of Oceanography, College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian 350108, China
- College of Chemical Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jing Jiang
- Fuzhou Institute of Oceanography, College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian 350108, China
| | - Gao Li
- Fuzhou Institute of Oceanography, College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian 350108, China
- Fujian-Taiwan-Hongkong-Macao Science and Technology Co-operation Base of Intelligent Pharmaceutics, Minjiang University, Fuzhou, Fujian 350108, China
| |
Collapse
|
7
|
Begum S, Parvej H, Dalui R, Paul S, Maity S, Sepay N, Afzal M, Chandra Halder U. Structural modulation of insulin by hydrophobic and hydrophilic molecules. RSC Adv 2023; 13:34097-34106. [PMID: 38019994 PMCID: PMC10662218 DOI: 10.1039/d3ra06647a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
In the bloodstream, insulin interacts with various kinds of molecules, which can alter its structure and modulate its function. In this work, we have synthesized two molecules having extremely hydrophilic and hydrophobic side chains. The effects of hydrophilic and hydrophobic molecules on the binding with insulin have been investigated through a multi-spectroscopic approach. We found that hydrophilic molecules have a slightly higher binding affinity towards insulin. Insulin can bind with the hydrophilic molecules as it binds glucose. The high insulin binding affinity of a hydrophobic molecule indicates its dual nature. The hydrophobic molecule binds at the hydrophobic pocket of the insulin surface, where hydrophilic molecules interact at the polar surface of the insulin. Such binding with the hydrophobic molecule perturbs strongly the secondary structure of the insulin much more in comparison to hydrophilic molecules. Therefore, the stability of insulin decreases in the presence of hydrophobic molecules.
Collapse
Affiliation(s)
- Shahnaz Begum
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Hasan Parvej
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Ramkrishna Dalui
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Swarnali Paul
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Sanhita Maity
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Nayim Sepay
- Department of Chemistry, Lady Brabourne College Kolkata-700017 India
| | - Mohd Afzal
- Department of Chemistry, College of Science, King Saud University Riyadh 11451 Saudi Arabia
| | | |
Collapse
|
8
|
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
|
9
|
Jia Y, Fernandez A, Sampath J. PEGylation of Insulin and Lysozyme To Stabilize against Thermal Denaturation: A Molecular Dynamics Simulation Study. J Phys Chem B 2023; 127:6856-6866. [PMID: 37498538 DOI: 10.1021/acs.jpcb.3c01289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Biologic drugs or "biologics" (proteins derived from living organisms) are one of the fastest-growing classes of FDA-approved therapeutics. These compounds are often fragile and require conjugation to polymers for stabilization, with many proteins too ephemeral for therapeutic use. During storage or administration, proteins tend to unravel and lose their secondary structure due to changes in solution temperature, pH, and other external stressors. To enhance their lifetime, protein drugs currently in the market are conjugated with polyethylene glycol (PEG), owing to its ability to increase the stability, solubility, and pharmacokinetics of protein drugs. Here, we perform all-atom molecular dynamics simulations to study the unfolding process of egg-white lysozyme and insulin at elevated temperatures. We test the validity of two force fields─CHARMM36 and Amber ff99SB-ILDN─in the unfolding process. By calculating global and local properties, we capture residues that deteriorate first─these are the "weak links" in the proteins. Next, we conjugate both proteins with PEG and find that PEG preserves the native structure of the proteins at elevated temperatures by blocking water molecules from entering the hydrophobic core, thereby causing the secondary structure to stabilize.
Collapse
Affiliation(s)
- Yinhao Jia
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Adam Fernandez
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Janani Sampath
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
10
|
Wang K, Chen K. Direct Assessment of Oligomerization of Chemically Modified Peptides and Proteins in Formulations using DLS and DOSY-NMR. Pharm Res 2023; 40:1329-1339. [PMID: 36627448 DOI: 10.1007/s11095-022-03468-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE Protein higher order structure (HOS) including the oligomer distribution can be critical for efficacy, safety and stability of drug products (DP). Oligomerization is particularly relevant to chemically modified protein therapeutics that have an extended pharmacokinetics profile. Therefore, the direct assessment of protein oligomerization in drug formulation is desired for quality assurance and control. METHODS Here, two non-invasive methods, dynamic light scattering (DLS) and diffusion ordered spectroscopy (DOSY) NMR, were applied to measure translational diffusion coefficients (Ddls and Dnmr) of proteins in formulated drug products. The hydrodynamic molecular weights (MWhd), similar to hydrodynamic size, of protein therapeutics were derived based on a log(Ddls) vs log(MWhd) correlation model established using protein standards. RESULTS An exponent value of -0.40 ± 0.01 was established for DLS measured log(D) vs. log(MWhd) using protein standards and a theoretical exponent value of -0.6 was used for unstructured polyethylene glycol (PEG) chains. The analysis of DLS derived MWhd of the primary species showed the fatty acid linked glucagon-like peptide 1 (GLP-1) was in different oligomer states, but the fatty acid linked insulin and PEG linked proteins were in monomer states. Nevertheless, equilibrium and exchange between oligomers in formulations were universal and clearly evidenced from DOSY-NMR for all drugs except peginterferon alfa-2a. CONCLUSION The correlation models of log(D) vs. log(MWhd) could be a quick and efficient way to predict MWhd of protein, which directly informs on the state of protein folding and oligomerization in formulation.
Collapse
Affiliation(s)
- Kai Wang
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Kang Chen
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA.
| |
Collapse
|
11
|
Curreri AM, Kim J, Dunne M, Angsantikul P, Goetz M, Gao Y, Mitragotri S. Deep Eutectic Solvents for Subcutaneous Delivery of Protein Therapeutics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205389. [PMID: 36642846 PMCID: PMC9982585 DOI: 10.1002/advs.202205389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/17/2022] [Indexed: 05/14/2023]
Abstract
Proteins are among the most common therapeutics for the treatment of diabetes, autoimmune diseases, cancer, and metabolic diseases, among others. Despite their common use, current protein therapies, most of which are injectables, have several limitations. Large proteins such as monoclonal antibodies (mAbs) suffer from poor absorption after subcutaneous injections, thus forcing their administration by intravenous injections. Even small proteins such as insulin suffer from slow pharmacokinetics which poses limitations in effective management of diabetes. Here, a deep eutectic-based delivery strategy is used to offer a generalized approach for improving protein absorption after subcutaneous injections. The lead formulation enhances absorption of mAbs after subcutaneous injections by ≈200%. The same composition also improves systemic absorption of subcutaneously injected insulin faster than Humalog, the current gold-standard of rapid acting insulin. Mechanistic studies reveal that the beneficial effect of deep eutectics on subcutaneous absorption is mediated by their ability to reduce the interactions of proteins with the subcutaneous matrix, especially collagen. Studies also confirm that these deep eutectics are safe for subcutaneous injections. Deep eutectic-based formulations described here open new possibilities for subcutaneous injections of therapeutic proteins.
Collapse
Affiliation(s)
- Alexander M. Curreri
- John A. Paulson School of Engineering and Applied SciencesHarvard University150 Western AveAllstonMA02134USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University3 Blackfan StBostonMA02115USA
| | - Jayoung Kim
- John A. Paulson School of Engineering and Applied SciencesHarvard University150 Western AveAllstonMA02134USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University3 Blackfan StBostonMA02115USA
| | - Michael Dunne
- John A. Paulson School of Engineering and Applied SciencesHarvard University150 Western AveAllstonMA02134USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University3 Blackfan StBostonMA02115USA
| | - Pavimol Angsantikul
- John A. Paulson School of Engineering and Applied SciencesHarvard University150 Western AveAllstonMA02134USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University3 Blackfan StBostonMA02115USA
- Present address:
The Population CouncilOne Dag Hammarskjold PlazaNew YorkNY10017USA
| | - Morgan Goetz
- John A. Paulson School of Engineering and Applied SciencesHarvard University150 Western AveAllstonMA02134USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University3 Blackfan StBostonMA02115USA
| | - Yongsheng Gao
- John A. Paulson School of Engineering and Applied SciencesHarvard University150 Western AveAllstonMA02134USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University3 Blackfan StBostonMA02115USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied SciencesHarvard University150 Western AveAllstonMA02134USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University3 Blackfan StBostonMA02115USA
| |
Collapse
|
12
|
Yang Y, Zhou R, Wang Y, Zhang Y, Yu J, Gu Z. Recent Advances in Oral and Transdermal Protein Delivery Systems. Angew Chem Int Ed Engl 2023; 62:e202214795. [PMID: 36478123 DOI: 10.1002/anie.202214795] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Protein and peptide drugs are predominantly administered by injection to achieve high bioavailability, but this greatly compromises patient compliance. Oral and transdermal drug delivery with minimal invasiveness and high adherence represent attractive alternatives to injection administration. However, oral and transdermal administration of bioactive proteins must overcome biological barriers, namely the gastrointestinal and skin barriers, respectively. The rapid development of new materials and technologies promises to address these physiological obstacles. This review provides an overview of the latest advances in oral and transdermal protein delivery, including chemical strategies, synthetic nanoparticles, medical microdevices, and biomimetic systems for oral administration, as well as chemical enhancers, physical approaches, and microneedles in transdermal delivery. We also discuss challenges and future perspectives of the field with a focus on innovation and translation.
Collapse
Affiliation(s)
- Yinxian Yang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ruyi Zhou
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yanfang Wang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jicheng Yu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.,Jinhua Institute of Zhejiang University, Jinhua, 321299, China.,Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhen Gu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.,Jinhua Institute of Zhejiang University, Jinhua, 321299, China.,Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
13
|
Clauss ZS, Meudom R, Su B, VandenBerg MA, Saini SS, Webber MJ, Chou DHC, Kramer JR. Supramolecular Protein Stabilization with Zwitterionic Polypeptide-Cucurbit[7]uril Conjugates. Biomacromolecules 2023; 24:481-488. [PMID: 36512327 DOI: 10.1021/acs.biomac.2c01319] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein aggregation is an obstacle for the development of new biopharmaceuticals, presenting challenges in shipping and storage of vital therapies. Though a variety of materials and methods have been explored, the need remains for a simple material that is biodegradable, nontoxic, and highly efficient at stabilizing protein therapeutics. In this work, we investigated zwitterionic polypeptides prepared using a rapid and scalable polymerization technique and conjugated to a supramolecular macrocycle host, cucurbit[7]uril, for the ability to inhibit aggregation of model protein therapeutics insulin and calcitonin. The polypeptides are based on the natural amino acid methionine, and zwitterion sulfonium modifications were compared to analogous cationic and neutral structures. Each polymer was end-modified with a single cucurbit[7]uril macrocycle to afford supramolecular recognition and binding to terminal aromatic amino acids on proteins. Only conjugates prepared from zwitterionic structures of sufficient chain lengths were efficient inhibitors of insulin aggregation and could also inhibit aggregation of calcitonin. This polypeptide exhibited no cytotoxicity in human cells even at concentrations that were five-fold of the intended therapeutic regime. We explored treatment of the zwitterionic polypeptides with a panel of natural proteases and found steady biodegradation as expected, supporting eventual clearance when used as a protein formulation additive.
Collapse
Affiliation(s)
- Zachary S Clauss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Rolande Meudom
- Department of Pediatrics, Division of Diabetes and Endocrinology, Stanford University, Palo Alto, California 94304, United States
| | - Bo Su
- Department of Chemical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Michael A VandenBerg
- Department of Chemical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Simranpreet S Saini
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Matthew J Webber
- Department of Chemical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Danny Hung-Chieh Chou
- Department of Pediatrics, Division of Diabetes and Endocrinology, Stanford University, Palo Alto, California 94304, United States
| | - Jessica R Kramer
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
14
|
Gelb M, Messina KMM, Vinciguerra D, Ko JH, Collins J, Tamboline M, Xu S, Ibarrondo FJ, Maynard HD. Poly(trehalose methacrylate) as an Excipient for Insulin Stabilization: Mechanism and Safety. ACS APPLIED MATERIALS & INTERFACES 2022; 14:37410-37423. [PMID: 35968684 PMCID: PMC9412841 DOI: 10.1021/acsami.2c09301] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/21/2022] [Indexed: 05/07/2023]
Abstract
Insulin, the oldest U.S. Food and Drug Administration (FDA)-approved recombinant protein and a World Health Organization (WHO) essential medicine for treating diabetes globally, faces challenges due to its storage instability. One approach to stabilize insulin is the addition of poly(trehalose methacrylate) (pTrMA) as an excipient. The polymer increases the stability of the peptide to heat and mechanical agitation and has a low viscosity suitable for injection and pumps. However, the safety and stabilizing mechanism of pTrMA is not yet known and is required to understand the potential suitability of pTrMA as an insulin excipient. Herein is reported the immune response, biodistribution, and insulin plasma lifetime in mice, as well as investigation into insulin stabilization. pTrMA alone or formulated with ovalbumin did not elicit an antibody response over 3 weeks in mice, and there was no observable cytokine production in response to pTrMA. Micropositron emission tomography/microcomputer tomography of 64Cu-labeled pTrMA showed excretion of 78-79% ID/cc within 24 h and minimal liver accumulation at 6-8% ID/cc when studied out to 120 h. Further, the plasma lifetime of insulin in mice was not altered by added pTrMA. Formulating insulin with 2 mol equiv of pTrMA improved the stability of insulin to standard storage conditions: 46 weeks at 4 °C yielded 87.0% intact insulin with pTrMA present as compared to 7.8% intact insulin without the polymer. The mechanism by which pTrMA-stabilized insulin was revealed to be a combination of inhibiting deamidation of amino acid residues and preventing fibrillation, followed by aggregation of inactive and immunogenic amyloids all without complexing insulin into its hexameric state, which could delay the onset of insulin activity. Based on the data reported here, we suggest that pTrMA stabilizes insulin as an excipient without adverse effects in vivo and is promising to investigate further for the safe formulation of insulin.
Collapse
Affiliation(s)
- Madeline
B. Gelb
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Kathryn M. M. Messina
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Daniele Vinciguerra
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Jeong Hoon Ko
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Jeffrey Collins
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine,
University of California, Los Angeles, California 90095-1735, United States
| | - Mikayla Tamboline
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine,
University of California, Los Angeles, California 90095-1735, United States
| | - Shili Xu
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine,
University of California, Los Angeles, California 90095-1735, United States
| | - F. Javier Ibarrondo
- Division
of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1569, United States
| | - Heather D. Maynard
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| |
Collapse
|
15
|
Maikawa CL, Mann JL, Kannan A, Meis CM, Grosskopf AK, Ou BS, Autzen AAA, Fuller GG, Maahs DM, Appel EA. Engineering Insulin Cold Chain Resilience to Improve Global Access. Biomacromolecules 2021; 22:3386-3395. [PMID: 34213889 PMCID: PMC8627795 DOI: 10.1021/acs.biomac.1c00474] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
There are 150 million people with diabetes worldwide who require insulin replacement therapy, and the prevalence of diabetes is rising the fastest in middle- and low-income countries. The current formulations require costly refrigerated transport and storage to prevent loss of insulin integrity. This study shows the development of simple "drop-in" amphiphilic copolymer excipients to maintain formulation integrity, bioactivity, pharmacokinetics, and pharmacodynamics for over 6 months when subjected to severe stressed aging conditions that cause current commercial formulation to fail in under 2 weeks. Further, when these copolymers are added to Humulin R (Eli Lilly) in original commercial packaging, they prevent insulin aggregation for up to 4 days at 50 °C compared to less than 1 day for Humulin R alone. These copolymers demonstrate promise as simple formulation additives to increase the cold chain resilience of commercial insulin formulations, thereby expanding global access to these critical drugs for treatment of diabetes.
Collapse
Affiliation(s)
- Caitlin L. Maikawa
- Department of Bioengineering, Stanford University, Stanford California 94305, United States
| | - Joseph L. Mann
- Department of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Aadithya Kannan
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Catherine M. Meis
- Department of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Abigail K. Grosskopf
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Ben S. Ou
- Department of Bioengineering, Stanford University, Stanford California 94305, United States
| | - Anton A. A. Autzen
- Department of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
- Department of Science and Technology, Aarhus University, Aarhus 8000, Denmark
| | - Gerald G. Fuller
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - David M. Maahs
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, California 94305, United States
- Diabetes Research Center, Stanford University, Stanford, California 94305, United States
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford California 94305, United States
- Department of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, California 94305, United States
- Diabetes Research Center, Stanford University, Stanford, California 94305, United States
- Stanford CHEM-H Institute, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
16
|
Maikawa CL, d’Aquino AI, Vuong ET, Su B, Zou L, Chen PC, Nguyen LT, Autzen AAA, Mann JL, Webber MJ, Appel EA. Affinity-Directed Dynamics of Host-Guest Motifs for Pharmacokinetic Modulation via Supramolecular PEGylation. Biomacromolecules 2021; 22:3565-3573. [PMID: 34314146 PMCID: PMC8627827 DOI: 10.1021/acs.biomac.1c00648] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Proteins are an impactful class of therapeutics but can exhibit suboptimal therapeutic performance, arising from poor control over the timescale of clearance. Covalent PEGylation is one established strategy to extend circulation time but often at the cost of reduced activity and increased immunogenicity. Supramolecular PEGylation may afford similar benefits without necessitating that the protein be permanently modified with a polymer. Here, we show that insulin pharmacokinetics can be modulated by tuning the affinity-directed dynamics of a host-guest motif used to non-covalently endow insulin with a poly(ethylene glycol) (PEG) chain. When administered subcutaneously, supramolecular PEGylation with higher binding affinities extends the time of total insulin exposure systemically. Pharmacokinetic modeling reveals that the extension in the duration of exposure arises specifically from decreased absorption from the subcutaneous depot governed directly by the affinity and dynamics of host-guest exchange. The lifetime of the supramolecular interaction thus dictates the rate of absorption, with negligible impact attributed to association of the PEG upon rapid dilution of the supramolecular complex in circulation. This modular approach to supramolecular PEGylation offers a powerful tool to tune protein pharmacokinetics in response to the needs of different disease applications.
Collapse
Affiliation(s)
- Caitlin L. Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Andrea I. d’Aquino
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Eric T. Vuong
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Bo Su
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Lei Zou
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Peyton C. Chen
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Leslee T. Nguyen
- Department of Biochemistry, Stanford University, Stanford, CA, 94305, USA
| | - Anton A. A. Autzen
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Science and Technology, Aarhus University, 8000 Aarhus, Denmark
| | - Joseph L. Mann
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Matthew J. Webber
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Endocrinology (Pediatrics), Stanford University, Stanford, CA, 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
17
|
Mann JL, Maikawa CL, Smith AAA, Grosskopf AK, Baker SW, Roth GA, Meis CM, Gale EC, Liong CS, Correa S, Chan D, Stapleton LM, Yu AC, Muir B, Howard S, Postma A, Appel EA. An ultrafast insulin formulation enabled by high-throughput screening of engineered polymeric excipients. Sci Transl Med 2021; 12:12/550/eaba6676. [PMID: 32611683 DOI: 10.1126/scitranslmed.aba6676] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
Insulin has been used to treat diabetes for almost 100 years; yet, current rapid-acting insulin formulations do not have sufficiently fast pharmacokinetics to maintain tight glycemic control at mealtimes. Dissociation of the insulin hexamer, the primary association state of insulin in rapid-acting formulations, is the rate-limiting step that leads to delayed onset and extended duration of action. A formulation of insulin monomers would more closely mimic endogenous postprandial insulin secretion, but monomeric insulin is unstable in solution using present formulation strategies and rapidly aggregates into amyloid fibrils. Here, we implement high-throughput-controlled radical polymerization techniques to generate a large library of acrylamide carrier/dopant copolymer (AC/DC) excipients designed to reduce insulin aggregation. Our top-performing AC/DC excipient candidate enabled the development of an ultrafast-absorbing insulin lispro (UFAL) formulation, which remains stable under stressed aging conditions for 25 ± 1 hours compared to 5 ± 2 hours for commercial fast-acting insulin lispro formulations (Humalog). In a porcine model of insulin-deficient diabetes, UFAL exhibited peak action at 9 ± 4 min, whereas commercial Humalog exhibited peak action at 25 ± 10 min. These ultrafast kinetics make UFAL a promising candidate for improving glucose control and reducing burden for patients with diabetes.
Collapse
Affiliation(s)
- Joseph L Mann
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Anton A A Smith
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA.,Department of Science and Technology, Aarhus University, 8000 Aarhus, Denmark
| | - Abigail K Grosskopf
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Sam W Baker
- Department of Comparative Medicine, Stanford University, Palo Alto, CA 94305, USA
| | - Gillie A Roth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Catherine M Meis
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Emily C Gale
- Department of Biochemistry, Stanford University, Palo Alto, CA 94305, USA
| | - Celine S Liong
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Santiago Correa
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Doreen Chan
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | | | - Anthony C Yu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Ben Muir
- CSIRO Manufacturing, Clayton, VIC 3168, Australia
| | - Shaun Howard
- CSIRO Manufacturing, Clayton, VIC 3168, Australia
| | - Almar Postma
- CSIRO Manufacturing, Clayton, VIC 3168, Australia
| | - Eric A Appel
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA. .,Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.,ChEM-H Institute, Stanford University, Stanford, CA 94305, USA.,Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
18
|
Bernhard S, Tibbitt MW. Supramolecular engineering of hydrogels for drug delivery. Adv Drug Deliv Rev 2021; 171:240-256. [PMID: 33561451 DOI: 10.1016/j.addr.2021.02.002] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
Supramolecular binding motifs are increasingly employed in the design of biomaterials. The ability to rationally engineer specific yet reversible associations into polymer networks with supramolecular chemistry enables injectable or sprayable hydrogels that can be applied via minimally invasive administration. In this review, we highlight two main areas where supramolecular binding motifs are being used in the design of drug delivery systems: engineering network mechanics and tailoring drug-material affinity. Throughout, we highlight many of the established and emerging chemistries or binding motifs that are useful for the design of supramolecular hydrogels for drug delivery applications.
Collapse
|
19
|
Maikawa CL, d'Aquino AI, Lal RA, Buckingham BA, Appel EA. Engineering biopharmaceutical formulations to improve diabetes management. Sci Transl Med 2021; 13:eabd6726. [PMID: 33504649 PMCID: PMC8004356 DOI: 10.1126/scitranslmed.abd6726] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
Insulin was first isolated almost a century ago, yet commercial formulations of insulin and its analogs for hormone replacement therapy still fall short of appropriately mimicking endogenous glycemic control. Moreover, the controlled delivery of complementary hormones (such as amylin or glucagon) is complicated by instability of the pharmacologic agents and complexity of maintaining multiple infusions. In this review, we highlight the advantages and limitations of recent advances in drug formulation that improve protein stability and pharmacokinetics, prolong drug delivery, or enable alternative dosage forms for the management of diabetes. With controlled delivery, these formulations could improve closed-loop glycemic control.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Andrea I d'Aquino
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Rayhan A Lal
- Department of Medicine (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Bruce A Buckingham
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
20
|
Liu Y, Zhang Y, Yu H, Liu Y. Cucurbituril‐Based Biomacromolecular Assemblies. Angew Chem Int Ed Engl 2020; 60:3870-3880. [DOI: 10.1002/anie.202009797] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Yao‐Hua Liu
- College of Chemistry State Key Laboratory of Elemento-Organic Chemistry Nankai University Tianjin 300071 China
| | - Ying‐Ming Zhang
- College of Chemistry State Key Laboratory of Elemento-Organic Chemistry Nankai University Tianjin 300071 China
| | - Hua‐Jiang Yu
- College of Chemistry State Key Laboratory of Elemento-Organic Chemistry Nankai University Tianjin 300071 China
| | - Yu Liu
- College of Chemistry State Key Laboratory of Elemento-Organic Chemistry Nankai University Tianjin 300071 China
| |
Collapse
|
21
|
Affiliation(s)
- Yao‐Hua Liu
- College of Chemistry State Key Laboratory of Elemento-Organic Chemistry Nankai University Tianjin 300071 China
| | - Ying‐Ming Zhang
- College of Chemistry State Key Laboratory of Elemento-Organic Chemistry Nankai University Tianjin 300071 China
| | - Hua‐Jiang Yu
- College of Chemistry State Key Laboratory of Elemento-Organic Chemistry Nankai University Tianjin 300071 China
| | - Yu Liu
- College of Chemistry State Key Laboratory of Elemento-Organic Chemistry Nankai University Tianjin 300071 China
| |
Collapse
|
22
|
Meis CM, Salzman EE, Maikawa CL, Smith AAA, Mann JL, Grosskopf AK, Appel EA. Self-Assembled, Dilution-Responsive Hydrogels for Enhanced Thermal Stability of Insulin Biopharmaceuticals. ACS Biomater Sci Eng 2020; 7:4221-4229. [PMID: 34510910 PMCID: PMC8441967 DOI: 10.1021/acsbiomaterials.0c01306] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
Biotherapeutics currently dominate
the landscape of new drugs because
of their exceptional potency and selectivity. Yet, the intricate molecular
structures that give rise to these beneficial qualities also render
them unstable in formulation. Hydrogels have shown potential as stabilizing
excipients for biotherapeutic drugs, providing protection against
harsh thermal conditions experienced during distribution and storage.
In this work, we report the utilization of a cellulose-based supramolecular
hydrogel formed from polymer–nanoparticle (PNP) interactions
to encapsulate and stabilize insulin, an important biotherapeutic
used widely to treat diabetes. Encapsulation of insulin in these hydrogels
prevents insulin aggregation and maintains insulin bioactivity through
stressed aging conditions of elevated temperature and continuous agitation
for over 28 days. Further, insulin can be easily recovered by dilution
of these hydrogels for administration at the point of care. This supramolecular
hydrogel system shows promise as a stabilizing excipient to reduce
the cold chain dependence of insulin and other biotherapeutics.
Collapse
Affiliation(s)
- Catherine M Meis
- Department of Materials Science & Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Erika E Salzman
- Department of Materials Science & Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Caitlin L Maikawa
- Department of Bioengineering, Stanford University, 443 Via Ortega, Stanford, California 94305, United States
| | - Anton A A Smith
- Department of Materials Science & Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States.,Department of Science and Technology, Aarhus University, 8000 Aarhus, Denmark
| | - Joseph L Mann
- Department of Materials Science & Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Abigail K Grosskopf
- Department of Chemical Engineering, Stanford University, 443 Via Ortega, Stanford, California 94305, United States
| | - Eric A Appel
- Department of Materials Science & Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States.,Department of Bioengineering, Stanford University, 443 Via Ortega, Stanford, California 94305, United States.,Department of Pediatrics-Endocrinology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, United States.,ChEM-H Institute, Stanford University, 290 Jane Stanford Way, Stanford, California 94305, United States
| |
Collapse
|
23
|
Quantitative analysis of weakly bound insulin oligomers in solution using polarized multidimensional fluorescence spectroscopy. Anal Chim Acta 2020; 1138:18-29. [PMID: 33161979 DOI: 10.1016/j.aca.2020.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 12/28/2022]
Abstract
Being able to measure the size and distribution of oligomers in solution is a critical issue in the manufacture and stability of insulin and other protein formulations. Measuring oligomers reliably can however be complicated, due to their fragile self-assembled structures, which are held together by weak forces. This can cause issues in chromatographic based methods, where dissociation or re-equilibration of oligomer populations can occur e.g. upon dilution in a different eluting buffer, but also for light scattering based methods like dynamic light scattering (DLS) where the size difference involved (often less than a factor 3) does not allow mixtures of oligomers to be resolved. Intrinsic fluorescence offers an attractive alternative as it is non-invasive, sensitive but also because it contains scattered light when implemented via excitation emission matrix (EEM) measurements, that is sensitive to changes in particle size. Here, using insulin at formulation level concentrations, we show for the first time how EEM can both discriminate and quantify the proportion of oligomeric states in solution. This was achieved by using the Rayleigh scatter (RS) band and the fluorescence signal contained in EEM. After validating size changes with DLS, we show in particular how the volume under the RS band correlated linearly with protein/oligomer molecular weight, in agreement with the Debye-Zimm relationship. This was true for the RS data from both EEM and polarized EEM (pEEM) measurements, the latter providing a stronger scatter signal, more sensitive to particle size changes. The fluorescence signal was then used with multivariate curve resolution (MCR) to quantify more precisely the soluble oligomer composition of insulin solutions. In conditions that promoted the formation of mainly one type of oligomer (monomer, dimer, or hexamer), pEEM-MCR helped identify the presence of small amounts of other oligomeric forms, while in conditions that were previously said to favour the insulin tetramer, we show that in the presence of zinc, these insulin samples were instead a heterogenous mixture composed of mostly dimers and hexamers. These MCR results correlated in all cases with the observed discrimination by principal component analysis (PCA), and deviations observed in the RS data. In conclusion, using pEEM scatter and emission components with chemometric data analysis provides a unique analytical method for characterising and monitoring changes in the soluble oligomeric state of proteins.
Collapse
|
24
|
A structurally minimized yet fully active insulin based on cone-snail venom insulin principles. Nat Struct Mol Biol 2020; 27:615-624. [PMID: 32483339 PMCID: PMC7374640 DOI: 10.1038/s41594-020-0430-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/07/2020] [Indexed: 12/26/2022]
Abstract
Human insulin and its current therapeutic analogs all show propensity, albeit varyingly, to self-associate into dimers and hexamers, which delays their onset of action and makes blood glucose management difficult for people with diabetes. Recently, we described a monomeric, insulin-like peptide in cone-snail venom with moderate human insulin-like bioactivity. Here, with insights from structural biology studies, we report the development of mini-Ins-a human des-octapeptide insulin analog-as a structurally minimal, full-potency insulin. Mini-Ins is monomeric and, despite the lack of the canonical B-chain C-terminal octapeptide, has similar receptor binding affinity to human insulin. Four mutations compensate for the lack of contacts normally made by the octapeptide. Mini-Ins also has similar in vitro insulin signaling and in vivo bioactivities to human insulin. The full bioactivity of mini-Ins demonstrates the dispensability of the PheB24-PheB25-TyrB26 aromatic triplet and opens a new direction for therapeutic insulin development.
Collapse
|
25
|
Smith AAA, Maikawa CL, Roth GA, Appel EA. Site-selective modification of proteins using cucurbit[7]uril as supramolecular protection for N-terminal aromatic amino acids. Org Biomol Chem 2020; 18:4371-4375. [PMID: 32459261 DOI: 10.1039/d0ob01004a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Cucurbit[7,8]urils are known to form inclusion complexes with aromatic amino acids, hosting the hydrohobic side chains within the cavity and adjacent cations within the portal of the macrocyclic host. Here we show that cucurbit[7]uril binding with N-terminal phenylalanine significantly reduces the nucleophilicity of the amine, likely due to an increase in stability of the ammonium ion, rendering it unreactive at neutral pH. Using insulin as a model protein, we show that this supramolecular protection strategy can drive selectivity of N-terminal amine conjugation away from the preferred B chain N-terminal phenylalanine towards the A chain N-terminal glycine. Cucurbit[7]uril can therefore be used as a supramolecular protecting group for site-selective protein modification.
Collapse
Affiliation(s)
- Anton A A Smith
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA. and Department of Chemistry, Aarhus University, 8000, Denmark
| | - Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Gillie A Roth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Eric A Appel
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA. and Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
26
|
Maikawa CL, Smith AAA, Zou L, Roth GA, Gale EC, Stapleton LM, Baker SW, Mann JL, Yu AC, Correa S, Grosskopf AK, Liong CS, Meis CM, Chan D, Troxell M, Maahs DM, Buckingham BA, Webber MJ, Appel EA. A co-formulation of supramolecularly stabilized insulin and pramlintide enhances mealtime glucagon suppression in diabetic pigs. Nat Biomed Eng 2020; 4:507-517. [PMID: 32393892 PMCID: PMC7274092 DOI: 10.1038/s41551-020-0555-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
Treatment of patients with diabetes with insulin and pramlintide (an amylin analogue) is more effective than treatment with insulin only. However, because mixtures of insulin and pramlintide are unstable and have to be injected separately, amylin analogues are only used by 1.5% of people with diabetes needing rapid-acting insulin. Here, we show that the supramolecular modification of insulin and pramlintide with cucurbit[7]uril-conjugated polyethylene glycol improves the pharmacokinetics of the dual-hormone therapy and enhances postprandial glucagon suppression in diabetic pigs. The co-formulation is stable for over 100 h at 37 °C under continuous agitation, whereas commercial formulations of insulin analogues aggregate after 10 h under similar conditions. In diabetic rats, the administration of the stabilized co-formulation increased the area-of-overlap ratio of the pharmacokinetic curves of pramlintide and insulin from 0.4 ± 0.2 to 0.7 ± 0.1 (mean ± s.d.) for the separate administration of the hormones. The co-administration of supramolecularly stabilized insulin and pramlintide better mimics the endogenous kinetics of co-secreted insulin and amylin, and holds promise as a dual-hormone replacement therapy.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Anton A A Smith
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Department of Science and Technology, Aarhus University, Aarhus, Denmark
| | - Lei Zou
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Gillie A Roth
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Emily C Gale
- Department of Biochemistry, Stanford University, Stanford, CA, USA
| | | | - Sam W Baker
- Department of Comparative Medicine, Stanford University, Stanford, CA, USA
| | - Joseph L Mann
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Anthony C Yu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Santiago Correa
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | | | - Celine S Liong
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Catherine M Meis
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Doreen Chan
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Megan Troxell
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - David M Maahs
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA
- Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Bruce A Buckingham
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA
- Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Matthew J Webber
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA.
- Diabetes Research Center, Stanford University, Stanford, CA, USA.
| |
Collapse
|