1
|
Chen W, Yu S, Webber B, DeWolf EL, Kilmer R, Xian S, Schmidt CR, Power EM, Webber MJ. Supramolecular Peptide Depots for Glucose-Responsive Glucagon Delivery. J Biomed Mater Res A 2025; 113:e37854. [PMID: 39719890 DOI: 10.1002/jbm.a.37854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024]
Abstract
Precise blood glucose control continues to be a critical challenge in the treatment and management of type 1 diabetes in order to mitigate both acute and chronic complications. This study investigates the development of a supramolecular peptide amphiphile (PA) material functionalized with phenylboronic acid (PBA) for glucose-responsive glucagon delivery. The PA-PBA system self-assembles into nanofibrillar hydrogels in the presence of physiological glucose levels, resulting in stable hydrogels capable of releasing glucagon under hypoglycemic conditions. Glucose responsiveness is driven by reversible binding between PBA and glucose, which modulates the electrostatic interactions necessary for hydrogel formation and dissolution. Through comprehensive in vitro characterization, including circular dichroism, zeta potential measurements, and rheological assessments, the PA-PBA system is found to exhibit glucose-dependent assembly, enabling controlled glucagon release that is inversely related to glucose concentration. Glucagon release is accelerated under low glucose conditions, simulating a hypoglycemic state, with a reduced rate seen at higher glucose levels. Evaluation of the platform in vivo using a type 1 diabetic mouse model demonstrates the efficacy in protecting against insulin-induced hypoglycemia by restoring blood glucose levels following an insulin overdose. The ability to tailor glucagon release in response to fluctuating glucose concentrations underscores the potential of this platform for improving glycemic control. These findings suggest that glucose-stabilized supramolecular peptide hydrogels hold significant promise for responsive drug delivery applications, offering an approach to manage glucose levels in diabetes and other metabolic disorders.
Collapse
Affiliation(s)
- Weike Chen
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Bernice Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Emily L DeWolf
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Rory Kilmer
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Sijie Xian
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Connor R Schmidt
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Elizabeth M Power
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| |
Collapse
|
2
|
Zhai C, Zulueta EC, Mariscal A, Xu C, Cui Y, Wang X, Wu H, Doan C, Wojtas L, Zhang H, Cai J, Ye L, Wang K, Liu W. From small changes to big gains: pyridinium-based tetralactam macrocycle for enhanced sugar recognition in water. Chem Sci 2024; 15:19588-19598. [PMID: 39568916 PMCID: PMC11575561 DOI: 10.1039/d4sc06190j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024] Open
Abstract
The complex distribution of functional groups in carbohydrates, coupled with their strong solvation in water, makes them challenging targets for synthetic receptors. Despite extensive research into various molecular frameworks, most synthetic carbohydrate receptors have exhibited low affinities, and their interactions with sugars in aqueous environments remain poorly understood. In this work, we present a simple pyridinium-based hydrogen-bonding receptor derived from a subtle structural modification of a well-known tetralactam macrocycle. This small structural change resulted in a dramatic enhancement of glucose binding affinity, increasing from 56 M-1 to 3001 M-1. Remarkably, the performance of our synthetic lectin surpasses that of the natural lectin, concanavalin A, by over fivefold. X-ray crystallography of the macrocycle-glucose complex reveals a distinctive hydrogen bonding pattern, which allows for a larger surface overlap between the receptor and glucose, contributing to the enhanced affinity. Furthermore, this receptor possesses allosteric binding sites, which involve chloride binding and trigger receptor aggregation. This unique allosteric process reveals the critical role of structural flexibility in this hydrogen-bonding receptor for the effective recognition of sugars. We also demonstrate the potential of this synthetic lectin as a highly sensitive glucose sensor in aqueous solutions.
Collapse
Affiliation(s)
- Canjia Zhai
- Department of Chemistry, University of South Florida Tampa FL 33620 USA
| | | | | | - Chengkai Xu
- Department of Chemistry, University of South Florida Tampa FL 33620 USA
| | - Yunpeng Cui
- Department of Chemistry, University of South Florida Tampa FL 33620 USA
| | - Xudong Wang
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida Tampa FL 33620 USA
| | - Huang Wu
- Department of Chemistry, The University of Hong Kong Hong Kong SAR 999077 China
| | - Carson Doan
- Department of Chemistry, University of South Florida Tampa FL 33620 USA
| | - Lukasz Wojtas
- Department of Chemistry, University of South Florida Tampa FL 33620 USA
| | - Haixin Zhang
- Department of Physics, University of Miami Coral Gables Florida 33146 USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida Tampa FL 33620 USA
| | - Libin Ye
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida Tampa FL 33620 USA
| | - Kun Wang
- Department of Physics, University of Miami Coral Gables Florida 33146 USA
- Department of Chemistry, University of Miami Coral Gables Florida 33146 USA
| | - Wenqi Liu
- Department of Chemistry, University of South Florida Tampa FL 33620 USA
| |
Collapse
|
3
|
Yu S, Webber MJ. Engineering disease analyte response in peptide self-assembly. J Mater Chem B 2024; 12:10757-10769. [PMID: 39382032 DOI: 10.1039/d4tb01860e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
A need to enhance the precision and specificity of therapeutic nanocarriers inspires the development of advanced nanomaterials capable of sensing and responding to disease-related cues. Self-assembled peptides offer a promising nanocarrier platform with versatile use to create precisely defined nanoscale materials. Disease-relevant cues can range from large biomolecules, such as enzymes, to ubiquitous small molecules with varying concentrations in healthy versus diseased states. Notably, pH changes (i.e., H+ concentration), redox species (e.g., H2O2), and glucose levels are significant spatial and/or temporal indicators of therapeutic need. Self-assembled peptides respond to these cues by altering their solubility, modulating electrostatic interactions, or facilitating chemical transformations through dynamic or labile bonds. This review explores the design and construction of therapeutic nanocarriers using self-assembled peptides, focusing on how peptide sequence engineering along with the inclusion of non-peptidic components can link the assembly state of these nanocarriers to the presence of disease-relevant small molecules.
Collapse
Affiliation(s)
- Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
4
|
Hoeg-Jensen T, Kruse T, Brand CL, Sturis J, Fledelius C, Nielsen PK, Nishimura E, Madsen AR, Lykke L, Halskov KS, Koščová S, Kotek V, Davis AP, Tromans RA, Tomsett M, Peñuelas-Haro G, Leonard DJ, Orchard MG, Chapman A, Invernizzi G, Johansson E, Granata D, Hansen BF, Pedersen TA, Kildegaard J, Pedersen KM, Refsgaard HHF, Alifrangis L, Fels JJ, Neutzsky-Wulff AV, Sauerberg P, Slaaby R. Glucose-sensitive insulin with attenuation of hypoglycaemia. Nature 2024; 634:944-951. [PMID: 39415004 PMCID: PMC11499270 DOI: 10.1038/s41586-024-08042-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/12/2024] [Indexed: 10/18/2024]
Abstract
The risk of inducing hypoglycaemia (low blood glucose) constitutes the main challenge associated with insulin therapy for diabetes1,2. Insulin doses must be adjusted to ensure that blood glucose values are within the normal range, but matching insulin doses to fluctuating glucose levels is difficult because even a slightly higher insulin dose than needed can lead to a hypoglycaemic incidence, which can be anything from uncomfortable to life-threatening. It has therefore been a long-standing goal to engineer a glucose-sensitive insulin that can auto-adjust its bioactivity in a reversible manner according to ambient glucose levels to ultimately achieve better glycaemic control while lowering the risk of hypoglycaemia3. Here we report the design and properties of NNC2215, an insulin conjugate with bioactivity that is reversibly responsive to a glucose range relevant for diabetes, as demonstrated in vitro and in vivo. NNC2215 was engineered by conjugating a glucose-binding macrocycle4 and a glucoside to insulin, thereby introducing a switch that can open and close in response to glucose and thereby equilibrate insulin between active and less-active conformations. The insulin receptor affinity for NNC2215 increased 3.2-fold when the glucose concentration was increased from 3 to 20 mM. In animal studies, the glucose-sensitive bioactivity of NNC2215 was demonstrated to lead to protection against hypoglycaemia while partially covering glucose excursions.
Collapse
Affiliation(s)
| | - Thomas Kruse
- Global Research Technologies, Novo Nordisk, Bagsværd, Denmark
| | | | - Jeppe Sturis
- Global Drug Discovery, Novo Nordisk, Bagsværd, Denmark
| | | | - Peter K Nielsen
- Global Research Technologies, Novo Nordisk, Bagsværd, Denmark
| | | | - Alice R Madsen
- Global Research Technologies, Novo Nordisk, Bagsværd, Denmark
| | - Lennart Lykke
- Global Research Technologies, Novo Nordisk, Bagsværd, Denmark
| | - Kim S Halskov
- Global Research Technologies, Novo Nordisk, Bagsværd, Denmark
| | | | | | | | | | | | | | | | | | | | | | - Eva Johansson
- Global Research Technologies, Novo Nordisk, Bagsværd, Denmark
| | - Daniele Granata
- Digital Science and Innovation, Novo Nordisk, Bagsværd, Denmark
| | - Bo F Hansen
- Global Drug Discovery, Novo Nordisk, Bagsværd, Denmark
| | | | | | | | | | | | - Johannes J Fels
- Global Research Technologies, Novo Nordisk, Bagsværd, Denmark
| | | | - Per Sauerberg
- Global Drug Discovery, Novo Nordisk, Bagsværd, Denmark
| | - Rita Slaaby
- Global Drug Discovery, Novo Nordisk, Bagsværd, Denmark.
| |
Collapse
|
5
|
Wang H, Bai S, Gu G, Zhang C, Wang Y. Chemical Reaction Steers Spatiotemporal Self-Assembly of Supramolecular Hydrogels. Chempluschem 2024; 89:e202400396. [PMID: 38923325 DOI: 10.1002/cplu.202400396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
Supramolecular structures are widespread in living system, which are usually spatiotemporally regulated by sophisticated metabolic processes to enable vital biological functions. Inspired by living system, tremendous efforts have been made to realize spatiotemporal control over the self-assembly of supramolecular materials in synthetic scenario by coupling chemical reaction with molecular self-assembly process. In this review, we focused on the works related to supramolecular hydrogels that are regulated in space and time using chemical reaction. Firstly, we summarized how spatially controlled self-assembly of supramolecular hydrogels can be achieved via chemical reaction-instructed self-assembly, and the application of such a self-assembly methodology in biotherapy was discussed as well. Second, we reviewed dynamic supramolecular hydrogels dictated by chemical reaction networks that can evolve their structures and properties against time. Third, we discussed the recent progresses in the control of the self-assembly of supramolecular hydrogels in both space and time though a reaction-diffusion-coupled self-assembly approach. Finally, we provided a perspective on the further development of spatiotemporally controlled supramolecular hydrogels using chemical reaction in the future.
Collapse
Affiliation(s)
- Hucheng Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Shengyu Bai
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Guanyao Gu
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Chunyu Zhang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yiming Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Shanghai Key Laboratory for Intelligent Sensing and Detection Technology, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
6
|
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
|
7
|
Zeng Z, Tan R, Chen S, Chen H, Liu Z, Liu L, Li M, Chen Y. Di-PEGylated insulin: A long-acting insulin conjugate with superior safety in reducing hypoglycemic events. Acta Pharm Sin B 2024; 14:2761-2772. [PMID: 38828152 PMCID: PMC11143505 DOI: 10.1016/j.apsb.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 06/05/2024] Open
Abstract
Although the discovery of insulin 100 years ago revolutionized the treatment of diabetes, its therapeutic potential is compromised by its short half-life and narrow therapeutic index. Current long-acting insulin analogs, such as insulin-polymer conjugates, are mainly used to improve pharmacokinetics by reducing renal clearance. However, these conjugates are synthesized without sacrificing the bioactivity of insulin, thus retaining the narrow therapeutic index of native insulin, and exceeding the efficacious dose still leads to hypoglycemia. Here, we report a kind of di-PEGylated insulin that can simultaneously reduce renal clearance and receptor-mediated clearance. By impairing the binding affinity to the receptor and the activation of the receptor, di-PEGylated insulin not only further prolongs the half-life of insulin compared to classical mono-PEGylated insulin but most importantly, increases its maximum tolerated dose 10-fold. The target of long-term glycemic management in vivo has been achieved through improved pharmacokinetics and a high dose. This work represents an essential step towards long-acting insulin medication with superior safety in reducing hypoglycemic events.
Collapse
Affiliation(s)
- Zhipeng Zeng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Runcheng Tan
- School of Materials Science and Engineering, Center of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Shi Chen
- School of Materials Science and Engineering, Center of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Haolin Chen
- School of Materials Science and Engineering, Center of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhijia Liu
- School of Materials Science and Engineering, Center of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Lixin Liu
- School of Materials Science and Engineering, Center of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yongming Chen
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- School of Materials Science and Engineering, Center of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
8
|
Bercea M, Lupu A. Recent Insights into Glucose-Responsive Concanavalin A-Based Smart Hydrogels for Controlled Insulin Delivery. Gels 2024; 10:260. [PMID: 38667679 PMCID: PMC11048858 DOI: 10.3390/gels10040260] [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: 02/25/2024] [Revised: 03/24/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Many efforts are continuously undertaken to develop glucose-sensitive biomaterials able of controlling glucose levels in the body and self-regulating insulin delivery. Hydrogels that swell or shrink as a function of the environmental free glucose content are suitable systems for monitoring blood glucose, delivering insulin doses adapted to the glucose concentration. In this context, the development of sensors based on reversible binding to glucose molecules represents a continuous challenge. Concanavalin A (Con A) is a bioactive protein isolated from sword bean plants (Canavalia ensiformis) and contains four sugar-binding sites. The high affinity for reversibly and specifically binding glucose and mannose makes Con A as a suitable natural receptor for the development of smart glucose-responsive materials. During the last few years, Con A was used to develop smart materials, such as hydrogels, microgels, nanoparticles and films, for producing glucose biosensors or drug delivery devices. This review is focused on Con A-based materials suitable in the diagnosis and therapeutics of diabetes. A brief outlook on glucose-derived theranostics of cancer is also presented.
Collapse
Affiliation(s)
- Maria Bercea
- “Petru Poni” Institute of Macromolecular Chemistry, 41-A Grigore Ghica Voda Alley, 700487 Iasi, Romania
| | - Alexandra Lupu
- “Petru Poni” Institute of Macromolecular Chemistry, 41-A Grigore Ghica Voda Alley, 700487 Iasi, Romania
| |
Collapse
|
9
|
Yu S, Ye Z, Roy R, Sonani RR, Pramudya I, Xian S, Xiang Y, Liu G, Flores B, Nativ-Roth E, Bitton R, Egelman EH, Webber MJ. Glucose-Triggered Gelation of Supramolecular Peptide Nanocoils with Glucose-Binding Motifs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311498. [PMID: 38095904 PMCID: PMC11031314 DOI: 10.1002/adma.202311498] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/11/2023] [Indexed: 12/28/2023]
Abstract
Peptide self-assembly is a powerful tool to prepare functional materials at the nanoscale. Often, the resulting materials have high aspect-ratio, with intermolecular β-sheet formation underlying 1D fibrillar structures. Inspired by dynamic structures in nature, peptide self-assembly is increasingly moving toward stimuli-responsive designs wherein assembled structures are formed, altered, or dissipated in response to a specific cue. Here, a peptide bearing a prosthetic glucose-binding phenylboronic acid (PBA) is demonstrated to self-assemble into an uncommon nanocoil morphology. These nanocoils arise from antiparallel β-sheets, with molecules aligned parallel to the long axis of the coil. The binding of glucose to the PBA motif stabilizes and elongates the nanocoil, driving entanglement and gelation at physiological glucose levels. The glucose-dependent gelation of these materials is then explored for the encapsulation and release of a therapeutic agent, glucagon, that corrects low blood glucose levels. Accordingly, the release of glucagon from the nanocoil hydrogels is inversely related to glucose level. When evaluated in a mouse model of severe acute hypoglycemia, glucagon delivered from glucose-stabilized nanocoil hydrogels demonstrates increased protection compared to delivery of the agent alone or within a control nanocoil hydrogel that is not stabilized by glucose.
Collapse
Affiliation(s)
- Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, 105 McCourtney Hall, Notre Dame, IN 46556, USA
| | - Zhou Ye
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, 105 McCourtney Hall, Notre Dame, IN 46556, USA
| | - Rajdip Roy
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, 105 McCourtney Hall, Notre Dame, IN 46556, USA
| | - Ravi R Sonani
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Irawan Pramudya
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, 105 McCourtney Hall, Notre Dame, IN 46556, USA
| | - Sijie Xian
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, 105 McCourtney Hall, Notre Dame, IN 46556, USA
| | - Yuanhui Xiang
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, 105 McCourtney Hall, Notre Dame, IN 46556, USA
| | - Guoqiang Liu
- Integrated Biomedical Sciences Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Belen Flores
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, 105 McCourtney Hall, Notre Dame, IN 46556, USA
| | - Einat Nativ-Roth
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Ronit Bitton
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, 105 McCourtney Hall, Notre Dame, IN 46556, USA
| |
Collapse
|
10
|
Xian S, Xiang Y, Liu D, Fan B, Mitrová K, Ollier RC, Su B, Alloosh MA, Jiráček J, Sturek M, Alloosh M, Webber MJ. Insulin-Dendrimer Nanocomplex for Multi-Day Glucose-Responsive Therapy in Mice and Swine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308965. [PMID: 37994248 DOI: 10.1002/adma.202308965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/27/2023] [Indexed: 11/24/2023]
Abstract
The management of diabetes in a manner offering autonomous insulin therapy responsive to glucose-directed need, and moreover with a dosing schedule amenable to facile administration, remains an ongoing goal to improve the standard of care. While basal insulins with reduced dosing frequency, even once-weekly administration, are on the horizon, there is still no approved therapy that offers glucose-responsive insulin function. Herein, a nanoscale complex combining both electrostatic- and dynamic-covalent interactions between a synthetic dendrimer carrier and an insulin analogue modified with a high-affinity glucose-binding motif yields an injectable insulin depot affording both glucose-directed and long-lasting insulin availability. Following a single injection, it is even possible to control blood glucose for at least one week in diabetic swine subjected to daily oral glucose challenges. Measurements of serum insulin concentration in response to challenge show increases in insulin corresponding to elevated blood glucose levels, an uncommon finding even in preclinical work on glucose-responsive insulin. Accordingly, the subcutaneous nanocomplex that results from combining electrostatic- and dynamic-covalent interactions between a modified insulin and a synthetic dendrimer carrier affords a glucose-responsive insulin depot for week-long control following a single routine injection.
Collapse
Affiliation(s)
- Sijie Xian
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Yuanhui Xiang
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Dongping Liu
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Bowen Fan
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Katarína Mitrová
- Czech Academy of Sciences, Institute of Organic Chemistry and Biochemistry, Prague, 16610, Czech Republic
| | - Rachel C Ollier
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Bo Su
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | | | - Jiří Jiráček
- Czech Academy of Sciences, Institute of Organic Chemistry and Biochemistry, Prague, 16610, Czech Republic
| | | | | | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| |
Collapse
|
11
|
Zhang Y, Tang Q, Zhou J, Zhao C, Li J, Wang H. Conductive and Eco-friendly Biomaterials-based Hydrogels for Noninvasive Epidermal Sensors: A Review. ACS Biomater Sci Eng 2024; 10:191-218. [PMID: 38052003 DOI: 10.1021/acsbiomaterials.3c01003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
As noninvasive wearable electronic devices, epidermal sensors enable continuous, real-time, and remote monitoring of various human physiological parameters. Conductive biomaterials-based hydrogels as sensor matrix materials have good biocompatibility, biodegradability, and efficient stimulus response capabilities and are widely applied in motion monitoring, healthcare, and human-machine interaction. However, biomass hydrogel-based epidermal sensing devices still need excellent mechanical properties, prolonged stability, multifunctionality, and extensive practicality. Therefore, this paper reviews the common biomass hydrogel materials for epidermal sensing (proteins, polysaccharides, polyphenols, etc.) and the various types of noninvasive sensing devices (strain/pressure sensors, temperature sensors, glucose sensors, electrocardiograms, etc.). Moreover, this review focuses on the strategies of scholars to enhance sensor properties, such as strength, conductivity, stability, adhesion, and self-healing ability. This work will guide the preparation and optimization of high-performance biomaterials-based hydrogel epidermal sensors.
Collapse
Affiliation(s)
- Yibo Zhang
- School of Information Science and Technology, Qingdao University of Science and Technology, Qingdao 266061, China
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, P. R. China
| | - Qianhui Tang
- School of Marine Technology and Environment, Dalian Ocean University, 52 Heishijiao Street, Dalian, Liaoning 116023, P. R. China
| | - Junyang Zhou
- School of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Chenghao Zhao
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, P. R. China
| | - Jingpeng Li
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, P. R. China
| | - Haiting Wang
- School of Information Science and Technology, Qingdao University of Science and Technology, Qingdao 266061, China
| |
Collapse
|
12
|
VandenBerg MA, Xian S, Xiang Y, Webber MJ. Dynamic-Covalent Crosslinking of Benzenetricarboxamide-Phenylboronate Conjugates. Macromol Biosci 2024; 24:e2300001. [PMID: 36786665 DOI: 10.1002/mabi.202300001] [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: 01/02/2023] [Revised: 02/06/2023] [Indexed: 02/15/2023]
Abstract
In an effort to augment the function of supramolecular biomaterials, recent efforts have explored the creation of hybrid materials that couple supramolecular and covalent components. Here, the benzenetricarboxamide (BTA) supramolecular polymer motif is modified to present a phenylboronic acid (PBA) in order to promote the crosslinking of 1D BTA stacks by PBA-diol dynamic-covalent bonds through the addition of a multi-arm diol-bearing crosslinker. Interestingly, the combination of these two motifs serves to frustrate the resulting assembly process, yielding hydrogels with worse mechanical properties than those prepared without the multi-arm diol crosslinker. Both systems with and without the crosslinker do, however, respond to the presence of a physiological level of glucose with a reduction in their mechanical integrity; repulsive electrostatic interactions in the BTA stacks occur in both cases upon glucose binding, with added competition from glucose with PBA-diol bonds amplifying glucose response in the hybrid material. Accordingly, the present results point to an unexpected outcome of reduced hydrogel mechanics, yet increased glucose response, when two disparate dynamic motifs of BTA supramolecular polymerization and PBA-diol crosslinking are combined, offering a vision for future preparation of glucose-responsive supramolecular biomaterials.
Collapse
Affiliation(s)
- Michael A VandenBerg
- Department of Chemical & Biomolecular Engineering, 205 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Sijie Xian
- Department of Chemical & Biomolecular Engineering, 205 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Yuanhui Xiang
- Department of Chemical & Biomolecular Engineering, 205 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, 205 McCourtney Hall, Notre Dame, IN, 46556, USA
| |
Collapse
|
13
|
Wang H, Mills J, Sun B, Cui H. Therapeutic Supramolecular Polymers: Designs and Applications. Prog Polym Sci 2024; 148:101769. [PMID: 38188703 PMCID: PMC10769153 DOI: 10.1016/j.progpolymsci.2023.101769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The self-assembly of low-molecular-weight building motifs into supramolecular polymers has unlocked a new realm of materials with distinct properties and tremendous potential for advancing medical practices. Leveraging the reversible and dynamic nature of non-covalent interactions, these supramolecular polymers exhibit inherent responsiveness to their microenvironment, physiological cues, and biomolecular signals, making them uniquely suited for diverse biomedical applications. In this review, we intend to explore the principles of design, synthesis methodologies, and strategic developments that underlie the creation of supramolecular polymers as carriers for therapeutics, contributing to the treatment and prevention of a spectrum of human diseases. We delve into the principles underlying monomer design, emphasizing the pivotal role of non-covalent interactions, directionality, and reversibility. Moreover, we explore the intricate balance between thermodynamics and kinetics in supramolecular polymerization, illuminating strategies for achieving controlled sizes and distributions. Categorically, we examine their exciting biomedical applications: individual polymers as discrete carriers for therapeutics, delving into their interactions with cells, and in vivo dynamics; and supramolecular polymeric hydrogels as injectable depots, with a focus on their roles in cancer immunotherapy, sustained drug release, and regenerative medicine. As the field continues to burgeon, harnessing the unique attributes of therapeutic supramolecular polymers holds the promise of transformative impacts across the biomedical landscape.
Collapse
Affiliation(s)
- Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jason Mills
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Boran Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
14
|
Yang JF, Yang S, Gong X, Bakh NA, Zhang G, Wang AB, Cherrington AD, Weiss MA, Strano MS. In Silico Investigation of the Clinical Translatability of Competitive Clearance Glucose-Responsive Insulins. ACS Pharmacol Transl Sci 2023; 6:1382-1395. [PMID: 37854621 PMCID: PMC10580396 DOI: 10.1021/acsptsci.3c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Indexed: 10/20/2023]
Abstract
The glucose-responsive insulin (GRI) MK-2640 from Merck was a pioneer in its class to enter the clinical stage, having demonstrated promising responsiveness in in vitro and preclinical studies via a novel competitive clearance mechanism (CCM). The smaller pharmacokinetic response in humans motivates the development of new predictive, computational tools that can improve the design of therapeutics such as GRIs. Herein, we develop and use a new computational model, IM3PACT, based on the intersection of human and animal model glucoregulatory systems, to investigate the clinical translatability of CCM GRIs based on existing preclinical and clinical data of MK-2640 and regular human insulin (RHI). Simulated multi-glycemic clamps not only validated the earlier hypothesis of insufficient glucose-responsive clearance capacity in humans but also uncovered an equally important mismatch between the in vivo competitiveness profile and the physiological glycemic range, which was not observed in animals. Removing the inter-species gap increases the glucose-dependent GRI clearance from 13.0% to beyond 20% for humans and up to 33.3% when both factors were corrected. The intrinsic clearance rate, potency, and distribution volume did not apparently compromise the translation. The analysis also confirms a responsive pharmacokinetics local to the liver. By scanning a large design space for CCM GRIs, we found that the mannose receptor physiology in humans remains limiting even for the most optimally designed candidate. Overall, we show that this computational approach is able to extract quantitative and mechanistic information of value from a posteriori analysis of preclinical and clinical data to assist future therapeutic discovery and development.
Collapse
Affiliation(s)
- Jing Fan Yang
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Sungyun Yang
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Xun Gong
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Naveed A. Bakh
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Ge Zhang
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Allison B. Wang
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Alan D. Cherrington
- Molecular
Physiology and Biophysics, Vanderbilt University
School of Medicine, Nashville, Tennessee 37232, United States
| | - Michael A. Weiss
- Department
of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Michael S. Strano
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
15
|
Zhang J, Chen F, Yu D, Liang Z, Dai F, Liang H, Li H, Tan H, Zhao L. Chitosan-based injectable hydrogels with dual glucose sensors for precise control of insulin release and diabetes mellitus therapy. Int J Pharm 2023; 643:123246. [PMID: 37467814 DOI: 10.1016/j.ijpharm.2023.123246] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/05/2023] [Accepted: 07/16/2023] [Indexed: 07/21/2023]
Abstract
Chitosan-based injectable hydrogels were designed and fabricated through the dynamic crosslinking of dual-reversible covalent bonds (imine and phenylboronate ester) for precise insulin release. The hydrogels contain dual glucose-sensors/responsive elements, featuring high sensitivity and rapid responsiveness to glucose level variation in cumulative and half-hourly pulsed insulin release. The hydrogels demonstrated improved cytocompatibility against HSF cells and histological long-term analysis of tissue after implantation. Evaluation of the glycemic control ability in STZ-induced hyperglycemic mice revealed that the hydrogel system showed excellent glycemic control ability in the glucose tolerance test and maintained blood glucose levels in a normal range for up to 11 days after a single administration. Thus, the hydrogel system showed applicable potential in insulin replacement therapy for diabetes mellitus.
Collapse
Affiliation(s)
- Jiaying Zhang
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 311215, China
| | - Fengjiao Chen
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 311215, China
| | - Dingle Yu
- Center for Child Care and Mental Health, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Zhenjiang Liang
- Center for Child Care and Mental Health, Shenzhen Children's Hospital, Shenzhen 518038, China.
| | - Fanjia Dai
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 311215, China
| | - Hongze Liang
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 311215, China
| | - Haiyan Li
- Department of Endocrinology, The First Affiliated Hospital, Shenzhen University, Shenzhen 518035, China
| | - Hui Tan
- Center for Child Care and Mental Health, Shenzhen Children's Hospital, Shenzhen 518038, China.
| | - Lingling Zhao
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 311215, China.
| |
Collapse
|
16
|
Herrera SE, Agazzi ML, Apuzzo E, Cortez ML, Marmisollé WA, Tagliazucchi M, Azzaroni O. Polyelectrolyte-multivalent molecule complexes: physicochemical properties and applications. SOFT MATTER 2023; 19:2013-2041. [PMID: 36811333 DOI: 10.1039/d2sm01507b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The complexation of polyelectrolytes with other oppositely charged structures gives rise to a great variety of functional materials with potential applications in a wide spectrum of technological fields. Depending on the assembly conditions, polyelectrolyte complexes can acquire different macroscopic configurations such as dense precipitates, nanosized colloids and liquid coacervates. In the past 50 years, much progress has been achieved to understand the principles behind the phase separation induced by the interaction of two oppositely charged polyelectrolytes in aqueous solutions, especially for symmetric systems (systems in which both polyions have similar molecular weight and concentration). However, in recent years, the complexation of polyelectrolytes with alternative building blocks such as small charged molecules (multivalent inorganic species, oligopeptides, and oligoamines, among others) has gained attention in different areas. In this review, we discuss the physicochemical characteristics of the complexes formed by polyelectrolytes and multivalent small molecules, putting a special emphasis on their similarities with the well-known polycation-polyanion complexes. In addition, we analyze the potential of these complexes to act as versatile functional platforms in various technological fields, such as biomedicine and advanced materials engineering.
Collapse
Affiliation(s)
- Santiago E Herrera
- Departamento de Química Inorgánica, Analítica y Química Física, INQUIMAE, CONICET. Facultad de Ciencias Exactas y Naturales. Ciudad Universitaria, Pabellón 2, Buenos Aires C1428EHA, Argentina.
| | - Maximiliano L Agazzi
- Instituto para el Desarrollo Agroindustrial y de la Salud (IDAS), (UNRC, CONICET), Ruta Nacional 36 KM 601, 5800 Río Cuarto, Argentina.
| | - Eugenia Apuzzo
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), (UNLP, CONICET), Sucursal 4, Casilla de Correo 16, 1900 La Plata, Argentina.
| | - M Lorena Cortez
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), (UNLP, CONICET), Sucursal 4, Casilla de Correo 16, 1900 La Plata, Argentina.
| | - Waldemar A Marmisollé
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), (UNLP, CONICET), Sucursal 4, Casilla de Correo 16, 1900 La Plata, Argentina.
| | - Mario Tagliazucchi
- Departamento de Química Inorgánica, Analítica y Química Física, INQUIMAE, CONICET. Facultad de Ciencias Exactas y Naturales. Ciudad Universitaria, Pabellón 2, Buenos Aires C1428EHA, Argentina.
| | - Omar Azzaroni
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), (UNLP, CONICET), Sucursal 4, Casilla de Correo 16, 1900 La Plata, Argentina.
| |
Collapse
|
17
|
Volpatti LR, Bochenek MA, Facklam AA, Burns DM, MacIsaac C, Morgart A, Walters B, Langer R, Anderson DG. Partially Oxidized Alginate as a Biodegradable Carrier for Glucose-Responsive Insulin Delivery and Islet Cell Replacement Therapy. Adv Healthc Mater 2023; 12:e2201822. [PMID: 36325648 PMCID: PMC9840661 DOI: 10.1002/adhm.202201822] [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: 07/27/2022] [Revised: 10/18/2022] [Indexed: 11/06/2022]
Abstract
Self-regulated insulin delivery that mimics native pancreas function has been a long-term goal for diabetes therapies. Two approaches towards this goal are glucose-responsive insulin delivery and islet cell transplantation therapy. Here, biodegradable, partially oxidized alginate carriers for glucose-responsive nanoparticles or islet cells are developed. Material composition and formulation are tuned in each of these contexts to enable glycemic control in diabetic mice. For injectable, glucose-responsive insulin delivery, 0.5 mm 2.5% oxidized alginate microgels facilitate repeat dosing and consistently provide 10 days of glycemic control. For islet cell transplantation, 1.5 mm capsules comprised of a blend of unoxidized and 2.5% oxidized alginate maintain cell viability and glycemic control over a period of more than 2 months while reducing the volume of nondegradable material implanted. These data show the potential of these biodegradable carriers for controlled drug and cell delivery for the treatment of diabetes with limited material accumulation in the event of multiple doses.
Collapse
Affiliation(s)
- Lisa R. Volpatti
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthew A. Bochenek
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Amanda A. Facklam
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Delaney M. Burns
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Corina MacIsaac
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexander Morgart
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Benjamin Walters
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G. Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
18
|
Shen D, Yu H, Wang L, Feng J, Zhang Q, Pan J, Han Y, Ni Z, Liang R, Uddin MA. Glucose-responsive nanoparticles designed via a molecular-docking-driven method for insulin delivery. J Control Release 2022; 352:527-539. [PMID: 36341933 DOI: 10.1016/j.jconrel.2022.10.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/21/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Nocturnal blood glucose regulation was one of the key challenges in diabetic treatments. However, development of the smart insulin complexes with mild and glucose-responsive delivering performances was mostly relied on experience of the senior researchers and numerous confirmation experiments. In this work, a series of bioinspired fatty-acid-modified glucose-responsive insulin-delivering polymeric nanoparticles were designed. The molecular docking technique was utilized to efficiently screen the fatty-acid-derived functional groups. The results provided the basis for polymer functionalization and simplified the optimization experiments. For the optimized formulation (C10MS), insulin-loaded C10MS successfully fulfilled the nocturnal-glycemic-controlling requirement of the diabetic rats with lower occurrence of hypoglycemia than the conventional insulin injection schemes. Such formulation also possessed good biocompatibility with the moderate elimination kinetics in vivo, which matched the demand of bio-safety in the daily treatments. Overall, this work opened up a new path for efficient design of functional polymeric materials.
Collapse
Affiliation(s)
- Di Shen
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Jingyi Feng
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Qian Zhang
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Jin Pan
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Yin Han
- Zhejiang Institute of Medical Device Testing, Hangzhou 310018, PR China
| | - Zhipeng Ni
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Ruixue Liang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Md Alim Uddin
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| |
Collapse
|
19
|
Xian S, VandenBerg MA, Xiang Y, Yu S, Webber MJ. Glucose-Responsive Injectable Thermogels via Dynamic-Covalent Cross-Linking of Pluronic Micelles. ACS Biomater Sci Eng 2022; 8:4873-4885. [PMID: 36317822 DOI: 10.1021/acsbiomaterials.2c00979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sijie Xian
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Michael A. VandenBerg
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Yuanhui Xiang
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Matthew J. Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
20
|
Hong Y, Yu H, Wang L, Chen X, Huang Y, Yang J, Ren S. Transdermal Insulin Delivery and Microneedles-based Minimally Invasive Delivery Systems. Curr Pharm Des 2022; 28:3175-3193. [PMID: 35676840 DOI: 10.2174/1381612828666220608130056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/25/2022] [Indexed: 01/28/2023]
Abstract
Diabetes has become a serious threat to human health, causing death and pain to numerous patients. Transdermal insulin delivery is a substitute for traditional insulin injection to avoid pain from the injection. Transdermal methods include non-invasive and invasive methods. As the non-invasive methods could hardly get through the stratum corneum, minimally invasive devices, especially microneedles, could enhance the transappendageal route in transcutaneous insulin delivery, and could act as connectors between the tissue and outer environment or devices. Microneedle patches have been in quick development in recent years and with different types, materials and functions. In those patches, the smart microneedle patch could perform as a sensor and reactor responding to glucose to regulate the blood level. In the smart microneedles field, the phenylboronic acid system and the glucose oxidase system have been successfully applied on the microneedle platform. Insulin transdermal delivery strategy, microneedles technology and smart microneedles' development would be discussed in this review.
Collapse
Affiliation(s)
- Yichuan Hong
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Xiang Chen
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Yudi Huang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Jian Yang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Shuning Ren
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| |
Collapse
|
21
|
Ye Z, Xiang Y, Monroe T, Yu S, Dong P, Xian S, Webber MJ. Polymeric Microneedle Arrays with Glucose-Sensing Dynamic-Covalent Bonding for Insulin Delivery. Biomacromolecules 2022; 23:4401-4411. [PMID: 36173091 DOI: 10.1021/acs.biomac.2c00878] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The ongoing rise in diabetes incidence necessitates improved therapeutic strategies to enable precise blood glucose control with convenient device form factors. Microneedle patches are one such device platform capable of achieving therapeutic delivery through the skin. In recent years, polymeric microneedle arrays have been reported using methods of in situ polymerization and covalent crosslinking in microneedle molds. In spite of promising results, in situ polymerization carries a risk of exposure to toxic unreacted precursors remaining in the device. Here, a polymeric microneedle patch is demonstrated that uses dynamic-covalent phenylboronic acid (PBA)-diol bonds in a dual role affording both network crosslinking and glucose sensing. By this approach, a pre-synthesized and purified polymer bearing pendant PBA motifs is combined with a multivalent diol crosslinker to prepare dynamic-covalent hydrogel networks. The ability of these dynamic hydrogels to shear-thin and self-heal enables their loading to a microneedle mold by centrifugation. Subsequent drying then yields a patch of uniformly shaped microneedles with the requisite mechanical properties to penetrate skin. Insulin release from these materials is accelerated in the presence of glucose. Moreover, short-term blood glucose control in a diabetic rat model following application of the device to the skin confirms insulin activity and bioavailability. Accordingly, dynamic-covalent crosslinking facilitates a route for fabricating microneedle arrays circumventing the toxicity concerns of in situ polymerization, offering a convenient device form factor for therapeutic insulin delivery.
Collapse
Affiliation(s)
- Zhou Ye
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556 United States
| | - Yuanhui Xiang
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556 United States
| | - Thomas Monroe
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556 United States
| | - Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556 United States
| | - Ping Dong
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556 United States
| | - Sijie Xian
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556 United States
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556 United States
| |
Collapse
|
22
|
Diboronate crosslinking: Introducing glucose specificity in glucose-responsive dynamic-covalent networks. J Control Release 2022; 348:601-611. [PMID: 35714732 DOI: 10.1016/j.jconrel.2022.06.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/24/2022] [Accepted: 06/10/2022] [Indexed: 11/21/2022]
Abstract
Dynamic-covalent motifs are increasingly used for hydrogel crosslinking, leveraging equilibrium-governed reversible bonds to prepare viscoelastic materials with dynamic properties and self-healing character. The bonding between aryl boronates and diols is one dynamic-covalent chemistry of interest. The extent of network crosslinking using this motif may be subject to competition from ambient diols such as glucose; this approach has long been explored for glucose-directed release of insulin to control diabetes. However, the majority of such work has used phenylboronic acids (PBAs) that suffer from low-affinity glucose binding, limiting material responsiveness. Moreover, many PBA chemistries also bind with higher affinity to certain non-glucose analytes like fructose and lactate than they do to glucose, limiting their specificity of sensing and therapeutic deployment. Here, dynamic-covalent hydrogels are prepared that, for the first time, use a new diboronate motif with enhanced glucose binding-and importantly improved glucose specificity-leveraging the ability of rigid diboronates to simultaneously bind two sites on a single glucose molecule. Compared to long-used PBA-based approaches, diboronate hydrogels offer more glucose-responsive insulin release that is minimally impacted by non-glucose analytes. Improved responsiveness translates to more rapid blood glucose correction in a rodent diabetes model. Accordingly, this new dynamic-covalent crosslinking chemistry is useful in realizing more sensitive and specific glucose-responsive materials.
Collapse
|
23
|
Huang D, Gao S, Luo Y, Zhou X, Lu Z, Zou L, Hu K, Zhao Z, Zhang Y. Glucose-sensitive membrane with phenylboronic acid-based contraction-type microgels as chemical valves. J Memb Sci 2022. [DOI: 10.1016/j.memsci.2022.120406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
24
|
Jarosinski MA, Chen YS, Varas N, Dhayalan B, Chatterjee D, Weiss MA. New Horizons: Next-Generation Insulin Analogues: Structural Principles and Clinical Goals. J Clin Endocrinol Metab 2022; 107:909-928. [PMID: 34850005 PMCID: PMC8947325 DOI: 10.1210/clinem/dgab849] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Indexed: 11/19/2022]
Abstract
Design of "first-generation" insulin analogues over the past 3 decades has provided pharmaceutical formulations with tailored pharmacokinetic (PK) and pharmacodynamic (PD) properties. Application of a molecular tool kit-integrating protein sequence, chemical modification, and formulation-has thus led to improved prandial and basal formulations for the treatment of diabetes mellitus. Although PK/PD changes were modest in relation to prior formulations of human and animal insulins, significant clinical advantages in efficacy (mean glycemia) and safety (rates of hypoglycemia) were obtained. Continuing innovation is providing further improvements to achieve ultrarapid and ultrabasal analogue formulations in an effort to reduce glycemic variability and optimize time in range. Beyond such PK/PD metrics, next-generation insulin analogues seek to exploit therapeutic mechanisms: glucose-responsive ("smart") analogues, pathway-specific ("biased") analogues, and organ-targeted analogues. Smart insulin analogues and delivery systems promise to mitigate hypoglycemic risk, a critical barrier to glycemic control, whereas biased and organ-targeted insulin analogues may better recapitulate physiologic hormonal regulation. In each therapeutic class considerations of cost and stability will affect use and global distribution. This review highlights structural principles underlying next-generation design efforts, their respective biological rationale, and potential clinical applications.
Collapse
Affiliation(s)
- Mark A Jarosinski
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nicolás Varas
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Deepak Chatterjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
25
|
Xu X, Ran Y, Huang C, Yin Z. Glucose and H 2O 2 Dual-Responsive Nanocomplex Grafted with Insulin Prodrug for Blood Glucose Regulation. Biomacromolecules 2022; 23:1765-1776. [PMID: 35275618 DOI: 10.1021/acs.biomac.2c00016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although "closed-loop" smart insulin delivery systems have been extensively investigated, the majority of them suffer from low insulin loading efficiency and slow glucose response. Here, we constructed a novel nanocomplex (NC), which was prepared by electrostatic interaction between negatively charged insulin prodrug nanoparticles (NPs) and positively charged polycaprolactone-polyethylenimine (PCL-PEI) micelles. The insulin prodrug was linked to acetalated dextran (AD) via borate ester bonds to form IAD NPs, and glucose oxidase (GOx) was encapsulated in PCL-PEI micelles. The NC was negatively charged with a high insulin grafting rate (0.473 mg/mg), and in vitro experiments revealed that IAD was sensitive to hyperglycemia and H2O2, whereas GOx significantly improved the response to glucose by altering the microenvironment to promote sustained insulin release. Furthermore, compared with free insulin and IAD NPs, subcutaneously injected NCs in diabetic rats had long-term hypoglycemic effects, showing excellent biocompatibility in vitro and in vivo, which had good potential in insulin self-regulation delivery.
Collapse
Affiliation(s)
- Xiaowen Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yu Ran
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chengyuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zongning Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
26
|
Liu Y, Zeng S, Ji W, Yao H, Lin L, Cui H, Santos HA, Pan G. Emerging Theranostic Nanomaterials in Diabetes and Its Complications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102466. [PMID: 34825525 PMCID: PMC8787437 DOI: 10.1002/advs.202102466] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/03/2021] [Indexed: 05/14/2023]
Abstract
Diabetes mellitus (DM) refers to a group of metabolic disorders that are characterized by hyperglycemia. Oral subcutaneously administered antidiabetic drugs such as insulin, glipalamide, and metformin can temporarily balance blood sugar levels, however, long-term administration of these therapies is associated with undesirable side effects on the kidney and liver. In addition, due to overproduction of reactive oxygen species and hyperglycemia-induced macrovascular system damage, diabetics have an increased risk of complications. Fortunately, recent advances in nanomaterials have provided new opportunities for diabetes therapy and diagnosis. This review provides a panoramic overview of the current nanomaterials for the detection of diabetic biomarkers and diabetes treatment. Apart from diabetic sensing mechanisms and antidiabetic activities, the applications of these bioengineered nanoparticles for preventing several diabetic complications are elucidated. This review provides an overall perspective in this field, including current challenges and future trends, which may be helpful in informing the development of novel nanomaterials with new functions and properties for diabetes diagnosis and therapy.
Collapse
Affiliation(s)
- Yuntao Liu
- School of Food & Biological EngineeringJiangsu UniversityZhenjiang212013China
- College of Food ScienceSichuan Agricultural UniversityYaan625014China
| | - Siqi Zeng
- College of Food ScienceSichuan Agricultural UniversityYaan625014China
| | - Wei Ji
- Department of PharmaceuticsSchool of PharmacyJiangsu UniversityZhenjiangJiangsu212013China
| | - Huan Yao
- Sichuan Institute of Food InspectionChengdu610097China
| | - Lin Lin
- School of Food & Biological EngineeringJiangsu UniversityZhenjiang212013China
| | - Haiying Cui
- School of Food & Biological EngineeringJiangsu UniversityZhenjiang212013China
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Department of Biomedical Engineering and W.J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity of Groningen/University Medical Center GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Guoqing Pan
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangJiangsu212013China
| |
Collapse
|
27
|
Yang L, Yang Y, Chen H, Mei L, Zeng X. Polymeric microneedle-mediated sustained release systems: Design strategies and promising applications for drug delivery. Asian J Pharm Sci 2022; 17:70-86. [PMID: 35261645 PMCID: PMC8888142 DOI: 10.1016/j.ajps.2021.07.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/24/2021] [Accepted: 07/03/2021] [Indexed: 12/24/2022] Open
Abstract
Parenteral sustained release drug formulations, acting as preferable platforms for long-term exposure therapy, have been wildly used in clinical practice. However, most of these delivery systems must be given by hypodermic injection. Therefore, issues including needle-phobic, needle-stick injuries and inappropriate reuse of needles would hamper the further applications of these delivery platforms. Microneedles (MNs) as a potential alternative system for hypodermic needles can benefit from minimally invasive and self-administration. Recently, polymeric microneedle-mediated sustained release systems (MN@SRS) have opened up a new way for treatment of many diseases. Here, we reviewed the recent researches in MN@SRS for transdermal delivery, and summed up its typical design strategies and applications in various diseases therapy, particularly focusing on the applications in contraception, infection, cancer, diabetes, and subcutaneous disease. An overview of the present clinical translation difficulties and future outlook of MN@SRS was also provided.
Collapse
Affiliation(s)
- Li Yang
- Institute of Pharmaceutics, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yao Yang
- Institute of Pharmaceutics, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Hongzhong Chen
- Institute of Pharmaceutics, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Lin Mei
- Institute of Pharmaceutics, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Xiaowei Zeng
- Institute of Pharmaceutics, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
28
|
Liu G, He S, Ding Y, Chen C, Cai Q, Zhou W. Multivesicular Liposomes for Glucose-Responsive Insulin Delivery. Pharmaceutics 2021; 14:21. [PMID: 35056918 PMCID: PMC8781467 DOI: 10.3390/pharmaceutics14010021] [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: 11/18/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
An intelligent insulin delivery system is highly desirable for diabetes management. Herein, we developed a novel glucose-responsive multivesicular liposome (MVL) for self-regulated insulin delivery using the double emulsion method. Glucose-responsive MVLs could effectively regulate insulin release in response to fluctuating glucose concentrations in vitro. Notably, in situ released glucose oxidase catalyzed glucose enrichment on the MVL surface, based on the combination of (3-fluoro-4-((octyloxy)carbonyl)phenyl)boronic acid and glucose. The outer MVL membrane was destroyed when triggered by the local acidic and H2O2-enriched microenvironment induced by glucose oxidase catalysis in situ, followed by the further release of entrapped insulin. Moreover, the Alizarin red probe and molecular docking were used to clarify the glucose-responsive mechanism of MVLs. Utilizing chemically induced type 1 diabetic rats, we demonstrated that the glucose-responsive MVLs could effectively regulate blood glucose levels within a normal range. Our findings suggest that glucose-responsive MVLs with good biocompatibility may have promising applications in diabetes treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, China; (G.L.); (S.H.); (Y.D.); (C.C.); (Q.C.)
| |
Collapse
|
29
|
|
30
|
Jarosinski MA, Dhayalan B, Chen YS, Chatterjee D, Varas N, Weiss MA. Structural principles of insulin formulation and analog design: A century of innovation. Mol Metab 2021; 52:101325. [PMID: 34428558 PMCID: PMC8513154 DOI: 10.1016/j.molmet.2021.101325] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The discovery of insulin in 1921 and its near-immediate clinical use initiated a century of innovation. Advances extended across a broad front, from the stabilization of animal insulin formulations to the frontiers of synthetic peptide chemistry, and in turn, from the advent of recombinant DNA manufacturing to structure-based protein analog design. In each case, a creative interplay was observed between pharmaceutical applications and then-emerging principles of protein science; indeed, translational objectives contributed to a growing molecular understanding of protein structure, aggregation and misfolding. SCOPE OF REVIEW Pioneering crystallographic analyses-beginning with Hodgkin's solving of the 2-Zn insulin hexamer-elucidated general features of protein self-assembly, including zinc coordination and the allosteric transmission of conformational change. Crystallization of insulin was exploited both as a step in manufacturing and as a means of obtaining protracted action. Forty years ago, the confluence of recombinant human insulin with techniques for site-directed mutagenesis initiated the present era of insulin analogs. Variant or modified insulins were developed that exhibit improved prandial or basal pharmacokinetic (PK) properties. Encouraged by clinical trials demonstrating the long-term importance of glycemic control, regimens based on such analogs sought to resemble daily patterns of endogenous β-cell secretion more closely, ideally with reduced risk of hypoglycemia. MAJOR CONCLUSIONS Next-generation insulin analog design seeks to explore new frontiers, including glucose-responsive insulins, organ-selective analogs and biased agonists tailored to address yet-unmet clinical needs. In the coming decade, we envision ever more powerful scientific synergies at the interface of structural biology, molecular physiology and therapeutics.
Collapse
Affiliation(s)
- Mark A Jarosinski
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Deepak Chatterjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Nicolás Varas
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA; Department of Chemistry, Indiana University, Bloomington, 47405, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, 47907, IN, USA.
| |
Collapse
|
31
|
Zhang J, Xu J, Lim J, Nolan JK, Lee H, Lee CH. Wearable Glucose Monitoring and Implantable Drug Delivery Systems for Diabetes Management. Adv Healthc Mater 2021; 10:e2100194. [PMID: 33930258 DOI: 10.1002/adhm.202100194] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Indexed: 12/11/2022]
Abstract
The global cost of diabetes care exceeds $1 trillion each year with more than $327 billion being spent in the United States alone. Despite some of the advances in diabetes care including continuous glucose monitoring systems and insulin pumps, the technology associated with managing diabetes has largely remained unchanged over the past several decades. With the rise of wearable electronics and novel functional materials, the field is well-poised for the next generation of closed-loop diabetes care. Wearable glucose sensors implanted within diverse platforms including skin or on-tooth tattoos, skin-mounted patches, eyeglasses, contact lenses, fabrics, mouthguards, and pacifiers have enabled noninvasive, unobtrusive, and real-time analysis of glucose excursions in ambulatory care settings. These wearable glucose sensors can be integrated with implantable drug delivery systems, including an insulin pump, glucose responsive insulin release implant, and islets transplantation, to form self-regulating closed-loop systems. This review article encompasses the emerging trends and latest innovations of wearable glucose monitoring and implantable insulin delivery technologies for diabetes management with a focus on their advanced materials and construction. Perspectives on the current unmet challenges of these strategies are also discussed to motivate future technological development toward improved patient care in diabetes management.
Collapse
Affiliation(s)
- Jinyuan Zhang
- Weldon School of Biomedical Engineering Purdue University West Lafayette IN 47907 USA
| | - Jian Xu
- Weldon School of Biomedical Engineering Purdue University West Lafayette IN 47907 USA
| | - Jongcheon Lim
- Weldon School of Biomedical Engineering Purdue University West Lafayette IN 47907 USA
| | - James K. Nolan
- Weldon School of Biomedical Engineering Purdue University West Lafayette IN 47907 USA
| | - Hyowon Lee
- Weldon School of Biomedical Engineering Purdue University West Lafayette IN 47907 USA
| | - Chi Hwan Lee
- Weldon School of Biomedical Engineering Purdue University West Lafayette IN 47907 USA
- School of Mechanical Engineering School of Materials Engineering Purdue University West Lafayette IN 47907 USA
| |
Collapse
|
32
|
Volpatti LR, Burns DM, Basu A, Langer R, Anderson DG. Engineered insulin-polycation complexes for glucose-responsive delivery with high insulin loading. J Control Release 2021; 338:71-79. [PMID: 34391834 DOI: 10.1016/j.jconrel.2021.08.017] [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: 03/01/2021] [Revised: 07/08/2021] [Accepted: 08/07/2021] [Indexed: 01/11/2023]
Abstract
Glucose-responsive insulin delivery systems have the potential to improve quality of life for individuals with diabetes by improving blood sugar control and limiting the risk of hypoglycemia. However, systems with desirable insulin release kinetics and high loading capacities have proven difficult to achieve. Here, we report the development of electrostatic complexes (ECs) comprised of insulin, a polycation, and glucose oxidase (GOx). Under normoglycemic physiological conditions, insulin carries a slight negative charge and forms a stable EC with the polycation. In hyperglycemia, the encapsulated glucose-sensing enzyme GOx converts glucose to gluconic acid and lowers the pH of the microenvironment, causing insulin to adopt a positive charge. Thus, the electrostatic interactions are disrupted, and insulin is released. Using a model polycation, we conducted molecular dynamics simulations to model these interactions, synthesized ECs with > 50% insulin loading capacity, and determined in vitro release kinetics. We further showed that a single dose of ECs can provide a glycemic profile in streptozotocin-induced diabetic mice that mimics healthy mice over a 9 h period with 2 glucose tolerance tests.
Collapse
Affiliation(s)
- Lisa R Volpatti
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Delaney M Burns
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arijit Basu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Anesthesiology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard-Massachusetts Institute of Technology, Division of Health Sciences and Technology, MA 02139, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Anesthesiology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard-Massachusetts Institute of Technology, Division of Health Sciences and Technology, MA 02139, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
33
|
Abstract
AbstractDiabetes is one of the most devastating global diseases with an ever-increasing number of patients. Achieving persistent glycemic control in a painless and convenient way is an unmet goal for diabetes management. Insulin therapy is commonly utilized for diabetes treatment and usually relies on patient self-injection. This not only impairs a patient’s quality of life and fails to precisely control the blood glucose level but also brings the risk of life-threatening hypoglycemia. “closed-loop” insulin delivery systems could avoid these issues by providing on-demand insulin delivery. However, safety concerns limit the application of currently developed electronics-derived or enzyme-based systems. Phenylboronic acid (PBA), with the ability to reversibly bind glucose and a chemically tailored binding specificity, has attracted substantial attention in recent years. This focus review provides an overview of PBA-based versatile insulin delivery platforms developed in our group, including new PBA derivatives, glucose-responsive gels, and gel-combined medical devices, with a unique “skin layer” controlled diffusion feature.
Collapse
|
34
|
Yu S, Xian S, Ye Z, Pramudya I, Webber MJ. Glucose-Fueled Peptide Assembly: Glucagon Delivery via Enzymatic Actuation. J Am Chem Soc 2021; 143:12578-12589. [PMID: 34280305 DOI: 10.1021/jacs.1c04570] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nature achieves remarkable function from the formation of transient, nonequilibrium materials realized through continuous energy input. The role of enzymes in catalyzing chemical transformations to drive such processes, often as part of stimuli-directed signaling, governs both material formation and lifetime. Inspired by the intricate nonequilibrium nanostructures of the living world, this work seeks to create transient materials in the presence of a consumable glucose stimulus under enzymatic control of glucose oxidase. Compared to traditional glucose-responsive materials, which typically engineer degradation to release insulin under high-glucose conditions, the transient nanofibrillar hydrogel materials here are stabilized in the presence of glucose but destabilized under conditions of limited glucose to release encapsulated glucagon. In the context of blood glucose control, glucagon offers a key antagonist to insulin in responding to hypoglycemia by signaling the release of glucose stored in tissues so as to restore normal blood glucose levels. Accordingly, these materials are evaluated in a prophylactic capacity in diabetic mice to release glucagon in response to a sudden drop in blood glucose brought on by an insulin overdose. Delivery of glucagon using glucose-fueled nanofibrillar hydrogels succeeds in limiting the onset and severity of hypoglycemia in mice. This general strategy points to a new paradigm in glucose-responsive materials, leveraging glucose as a stabilizing cue for responsive glucagon delivery in combating hypoglycemia. Moreover, compared to most fundamental reports achieving nonequilibrium and/or fueled classes of materials, the present work offers a rare functional example using a disease-relevant fuel to drive deployment of a therapeutic.
Collapse
Affiliation(s)
- Sihan Yu
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, Indiana 46556, United States
| | - Sijie Xian
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, Indiana 46556, United States
| | - Zhou Ye
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, Indiana 46556, United States
| | - Irawan Pramudya
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, Indiana 46556, United States
| | - Matthew J Webber
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, Indiana 46556, United States
| |
Collapse
|
35
|
Xu L, Zhang X, Wang Z, Haidry AA, Yao Z, Haque E, Wang Y, Li G, Daeneke T, McConville CF, Kalantar-Zadeh K, Zavabeti A. Low dimensional materials for glucose sensing. NANOSCALE 2021; 13:11017-11040. [PMID: 34152349 DOI: 10.1039/d1nr02529e] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Biosensors are essential components for effective healthcare management. Since biological processes occur on molecular scales, nanomaterials and nanosensors intrinsically provide the most appropriate landscapes for developing biosensors. Low-dimensional materials have the advantage of offering high surface areas, increased reactivity and unique physicochemical properties for efficient and selective biosensing. So far, nanomaterials and nanodevices have offered significant prospects for glucose sensing. Targeted glucose biosensing using such low-dimensional materials enables much more effective monitoring of blood glucose levels, thus providing significantly better predictive diabetes diagnostics and management. In this review, recent advances in using low dimensional materials for sensing glucose are summarized. Sensing fundamentals are discussed, as well as invasive, minimally-invasive and non-invasive sensing methods. The effects of morphological characteristics and size-dependent properties of low dimensional materials are explored for glucose sensing, and the key performance parameters such as selectivity, stability and sensitivity are also discussed. Finally, the challenges and future opportunities that low dimensional materials can offer for glucose sensing are outlined.
Collapse
Affiliation(s)
- Linling Xu
- College of Materials Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211100, China
| | - Xianfei Zhang
- College of Materials Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211100, China
| | - Zhe Wang
- College of Materials Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211100, China
| | - Azhar Ali Haidry
- College of Materials Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211100, China
| | - Zhengjun Yao
- College of Materials Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211100, China
| | - Enamul Haque
- School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
| | - Yichao Wang
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia
| | - Gang Li
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, 3010 Australia.
| | - Torben Daeneke
- School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
| | - Chris F McConville
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia
| | - Kourosh Kalantar-Zadeh
- School of Chemical Engineering, University of New South Wales (UNSW), Kensington, NSW 2052, Australia.
| | - Ali Zavabeti
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, 3010 Australia.
| |
Collapse
|
36
|
Wang Y, Wang H, Zhu XX, Guan Y, Zhang Y. Smart microneedle patches for rapid, and painless transdermal insulin delivery. J Mater Chem B 2021; 8:9335-9342. [PMID: 32969458 DOI: 10.1039/d0tb01822h] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Insulin administration at mealtimes for the control of postprandial glucose is a major part of basal-bolus insulin therapy; however, painful subcutaneous (SC) injections lead to poor patient compliance. The microneedle (MN) patch, which allows painless transdermal drug delivery, is a promising substitute; however, it remains a big challenge to deliver insulin as rapidly as by SC injection. Here a novel MN patch is designed in which the MNs are coated with insulin/poly-l-glutamic acid (PGA) layer-by-layer (LBL) films at pH 3.0. This coating is pH-sensitive because the net charge of insulin turns from positive to negative when the pH increases from 3.0 to 7.4. As a result, when transferred to pH 7.4 media, e.g., when inserted into skin, the coating dissociates instantly and releases insulin rapidly. A brief epidermal application (<1 min) of the coated MNs is enough for complete film dissociation. More importantly, the coated MN patch exhibits a pharmacokinetic and a pharmacodynamic profile comparable to that of insulin administrated by SC injection, suggesting the coated MN patch can deliver insulin as rapidly as the SC injection. In addition, the patch exhibits excellent biocompatibility and storage stability. The new MN patch is expected to become a painless, convenient method for the control of postprandial glucose.
Collapse
Affiliation(s)
- Yuanpeng Wang
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Haozheng Wang
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - X X Zhu
- Département de Chimie, Université de Montréal, C.P. 6128, Succ. Centre-ville, Montreal, QC H3C 3J7, Canada
| | - Ying Guan
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Yongjun Zhang
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| |
Collapse
|
37
|
Abstract
BACKGROUND Hypoglycemia, the condition of low blood sugar, is a common occurance in people with diabetes using insulin therapy. Protecting against hypoglycaemia by engineering an insulin preparation that can auto-adjust its biological activity to fluctuating blood glucose levels has been pursued since the 1970s, but despite numerous publications, no system that works well enough for practical use has reached clinical practise. SCOPE OF REVIEW This review will summarise and scrutinise known approaches for producing glucose-sensitive insulin therapies. Notably, systems described in patent applications will be extensively covered, which has not been the case for earlier reviews of this area. MAJOR CONCLUSIONS The vast majority of published systems are not suitable for product development, but a few glucose-sensitive insulin concepts have recently reached clinical trials, and there is hope that glucose-sensitive insulin will become available to people with diabetes in the near future.
Collapse
Affiliation(s)
- Thomas Hoeg-Jensen
- Research Chemistry, Novo Nordisk A/S, Novo Nordisk Park H5.S.05, DK-2720 Maaloev, Denmark.
| |
Collapse
|
38
|
Wang J, Wang Z, Chen G, Wang Y, Ci T, Li H, Liu X, Zhou D, Kahkoska AR, Zhou Z, Meng H, Buse JB, Gu Z. Injectable Biodegradable Polymeric Complex for Glucose-Responsive Insulin Delivery. ACS NANO 2021; 15:4294-4304. [PMID: 33685124 PMCID: PMC8210813 DOI: 10.1021/acsnano.0c07291] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Insulin therapy is the central component of treatment for type 1 and advanced type 2 diabetes; however, its narrow therapeutic window is associated with a risk of severe hypoglycemia. A glucose-responsive carrier that demonstrates consistent and slow basal insulin release under a normoglycemic condition and accelerated insulin release in response to hyperglycemia in real-time could offer effective blood glucose regulation with reduced risk of hypoglycemia. Here, we describe a poly(l-lysine)-derived biodegradable glucose-responsive cationic polymer for constructing polymer-insulin complexes for glucose-stimulated insulin delivery. The effects of the modification degree of arylboronic acid in the synthesized cationic polymer and polymer-to-insulin ratio on the glucose-dependent equilibrated free insulin level and the associated insulin release kinetics have been studied. In addition, the blood glucose regulation ability of these complexes and the associated glucose challenge-triggered insulin release are evaluated in type 1 diabetic mice.
Collapse
Affiliation(s)
- Jinqiang Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Guojun Chen
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Department of Biomedical Engineering, and the Rosalind & Morris Goodman Cancer Research Center, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Yanfang Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Ci
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Hongjun Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiangsheng Liu
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine, Los Angeles, California 90095, United States
| | - Daojia Zhou
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Anna R Kahkoska
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Zhuxian Zhou
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Huan Meng
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90024, United States
| | - John B Buse
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90024, United States
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Zhejiang Laboratory of Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
| |
Collapse
|
39
|
Zhang T, Tang JZ, Fei X, Li Y, Song Y, Qian Z, Peng Q. Can nanoparticles and nano‒protein interactions bring a bright future for insulin delivery? Acta Pharm Sin B 2021; 11:651-667. [PMID: 33777673 PMCID: PMC7982494 DOI: 10.1016/j.apsb.2020.08.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 02/05/2023] Open
Abstract
Insulin therapy plays an essential role in the treatment of diabetes mellitus. However, frequent injections required to effectively control the glycemic levels lead to substantial inconvenience and low patient compliance. In order to improve insulin delivery, many efforts have been made, such as developing the nanoparticles (NPs)-based release systems and oral insulin. Although some improvements have been achieved, the ultimate results are still unsatisfying and none of insulin-loaded NPs systems have been approved for clinical use so far. Recently, nano‒protein interactions and protein corona formation have drawn much attention due to their negative influence on the in vivo fate of NPs systems. As the other side of a coin, such interactions can also be used for constructing advanced drug delivery systems. Herein, we aim to provide an insight into the advance and flaws of various NPs-based insulin delivery systems. Particularly, an interesting discussion on nano‒protein interactions and its potentials for developing novel insulin delivery systems is initiated. Insulin therapy plays essential roles in treating diabetes. Optimizing insulin delivery enhances insulin therapy. Nanoparticles are promising systems for delivery of insulin. Nano-protein interactions influence the delivery of nanoparticles. Nano-protein interactions can be used for advanced delivery of insulin.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - James Zhenggui Tang
- Research Institute in Healthcare Science, Faculty of Science and Engineering, School of Pharmacy, University of Wolverhampton, Wolverhampton, WV1 1LY, UK
| | - Xiaofan Fei
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yi Song
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Corresponding author.
| |
Collapse
|
40
|
Wang Z, Wang J, Kahkoska AR, Buse JB, Gu Z. Developing Insulin Delivery Devices with Glucose Responsiveness. Trends Pharmacol Sci 2021; 42:31-44. [PMID: 33250274 PMCID: PMC7758938 DOI: 10.1016/j.tips.2020.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022]
Abstract
Individuals with type 1 and advanced type 2 diabetes require daily insulin therapy to maintain blood glucose levels in normoglycemic ranges to prevent associated morbidity and mortality. Optimal insulin delivery should offer both precise dosing in response to real-time blood glucose levels as well as a feasible and low-burden administration route to promote long-term adherence. A series of glucose-responsive insulin delivery mechanisms and devices have been reported to increase patient compliance while mitigating the risk of hypoglycemia. This review discusses currently available insulin delivery devices, overviews recent developments towards the generation of glucose-responsive delivery systems, and provides commentary on the opportunities and barriers ahead regarding the integration and translation of current glucose-responsive insulin delivery designs.
Collapse
Affiliation(s)
- Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Jinqiang Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA; College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Anna R Kahkoska
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - John B Buse
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA; College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China; California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
41
|
Abstract
Insulin therapy has advanced remarkably over the past few decades. Ultra-rapid-acting and ultra-long-acting insulin analogs are now commercially available. Many additional insulin formulations are in development. This review outlines recent advances in insulin therapy and novel therapies in development.
Collapse
Affiliation(s)
- Leah M. Wilson
- Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Jessica R. Castle
- Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, Portland, Oregon, USA
- Address correspondence to: Jessica R. Castle, MD, Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, L607, Portland, OR 97239-3098, USA
| |
Collapse
|
42
|
Microgel encapsulated nanoparticles for glucose-responsive insulin delivery. Biomaterials 2020; 267:120458. [PMID: 33197650 DOI: 10.1016/j.biomaterials.2020.120458] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 01/12/2023]
Abstract
An insulin delivery system that self-regulates blood glucose levels has the potential to limit hypoglycemic events and improve glycemic control. Glucose-responsive insulin delivery systems have been developed by coupling glucose oxidase with a stimuli-responsive biomaterial. However, the challenge of achieving desirable release kinetics (i.e., insulin release within minutes after glucose elevation and duration of release on the order of weeks) still remains. Here, we develop a glucose-responsive delivery system using encapsulated glucose-responsive, acetalated-dextran nanoparticles in porous alginate microgels. The nanoparticles respond rapidly to changes in glucose concentrations while the microgels provide them with protection and stability, allowing for extended glucose-responsive insulin release. This system reduces blood sugar in a diabetic mouse model at a rate similar to naked insulin and responds to a glucose challenge 3 days after administration similarly to a healthy animal. With 2 doses of microgels containing 60 IU/kg insulin each, we are able to achieve extended glycemic control in diabetic mice for 22 days.
Collapse
|
43
|
Yang JF, Gong X, Bakh NA, Carr K, Phillips NFB, Ismail-Beigi F, Weiss MA, Strano MS. Connecting Rodent and Human Pharmacokinetic Models for the Design and Translation of Glucose-Responsive Insulin. Diabetes 2020; 69:1815-1826. [PMID: 32152206 PMCID: PMC8176262 DOI: 10.2337/db19-0879] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/08/2020] [Indexed: 12/16/2022]
Abstract
Despite considerable progress, development of glucose-responsive insulins (GRIs) still largely depends on empirical knowledge and tedious experimentation-especially on rodents. To assist the rational design and clinical translation of the therapeutic, we present a Pharmacokinetic Algorithm Mapping GRI Efficacies in Rodents and Humans (PAMERAH) built upon our previous human model. PAMERAH constitutes a framework for predicting the therapeutic efficacy of a GRI candidate from its user-specified mechanism of action, kinetics, and dosage, which we show is accurate when checked against data from experiments and literature. Results from simulated glucose clamps also agree quantitatively with recent GRI publications. We demonstrate that the model can be used to explore the vast number of permutations constituting the GRI parameter space and thereby identify the optimal design ranges that yield desired performance. A design guide aside, PAMERAH more importantly can facilitate GRI's clinical translation by connecting each candidate's efficacies in rats, mice, and humans. The resultant mapping helps to find GRIs that appear promising in rodents but underperform in humans (i.e., false positives). Conversely, it also allows for the discovery of optimal human GRI dynamics not captured by experiments on a rodent population (false negatives). We condense such information onto a "translatability grid" as a straightforward, visual guide for GRI development.
Collapse
Affiliation(s)
- Jing Fan Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Xun Gong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Naveed A Bakh
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Kelley Carr
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH
| | | | | | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Michael S Strano
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
44
|
Nanoparticles-encapsulated polymeric microneedles for transdermal drug delivery. J Control Release 2020; 325:163-175. [PMID: 32629134 DOI: 10.1016/j.jconrel.2020.06.039] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/20/2022]
Abstract
Polymeric microneedles (MNs) have been leveraged as a novel transdermal drug delivery platform for effective drug permeation, which were widely used in the treatment of various diseases. However, issues including limited loading capacity of hydrophobic drugs, uncontrollable drug release rates, and monotonic therapeutic strategy hamper the further application of polymeric MNs. As a recent emerging research topic, drawing inspiration from the ways that nanomedicine integrated with MNs have opened new avenues for disease therapy. In this review, we examined the recent studies employing nanoparticles (NPs)-encapsulated polymeric MNs (NPs@MNs) for transdermal delivery of various therapeutic cargos, particularly focused on the application of NPs@MNs for diabetes therapy, infectious disease therapy, cancer therapy, and other dermatological disease therapy. We also provided an overview of the clinical potential and future translation of NPs@MNs.
Collapse
|
45
|
Das SS, Bharadwaj P, Bilal M, Barani M, Rahdar A, Taboada P, Bungau S, Kyzas GZ. Stimuli-Responsive Polymeric Nanocarriers for Drug Delivery, Imaging, and Theragnosis. Polymers (Basel) 2020; 12:E1397. [PMID: 32580366 PMCID: PMC7362228 DOI: 10.3390/polym12061397] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
In the past few decades, polymeric nanocarriers have been recognized as promising tools and have gained attention from researchers for their potential to efficiently deliver bioactive compounds, including drugs, proteins, genes, nucleic acids, etc., in pharmaceutical and biomedical applications. Remarkably, these polymeric nanocarriers could be further modified as stimuli-responsive systems based on the mechanism of triggered release, i.e., response to a specific stimulus, either endogenous (pH, enzymes, temperature, redox values, hypoxia, glucose levels) or exogenous (light, magnetism, ultrasound, electrical pulses) for the effective biodistribution and controlled release of drugs or genes at specific sites. Various nanoparticles (NPs) have been functionalized and used as templates for imaging systems in the form of metallic NPs, dendrimers, polymeric NPs, quantum dots, and liposomes. The use of polymeric nanocarriers for imaging and to deliver active compounds has attracted considerable interest in various cancer therapy fields. So-called smart nanopolymer systems are built to respond to certain stimuli such as temperature, pH, light intensity and wavelength, and electrical, magnetic and ultrasonic fields. Many imaging techniques have been explored including optical imaging, magnetic resonance imaging (MRI), nuclear imaging, ultrasound, photoacoustic imaging (PAI), single photon emission computed tomography (SPECT), and positron emission tomography (PET). This review reports on the most recent developments in imaging methods by analyzing examples of smart nanopolymers that can be imaged using one or more imaging techniques. Unique features, including nontoxicity, water solubility, biocompatibility, and the presence of multiple functional groups, designate polymeric nanocues as attractive nanomedicine candidates. In this context, we summarize various classes of multifunctional, polymeric, nano-sized formulations such as liposomes, micelles, nanogels, and dendrimers.
Collapse
Affiliation(s)
- Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India;
| | - Priyanshu Bharadwaj
- UFR des Sciences de Santé, Université de Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
| | - Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 76175-133, Iran;
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Pablo Taboada
- Colloids and Polymers Physics Group, Condensed Matter Physics Area, Particle Physics Department Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - George Z. Kyzas
- Department of Chemistry, International Hellenic University, 65404 Kavala, Greece
| |
Collapse
|
46
|
Schiller JL, Lai SK. Tuning Barrier Properties of Biological Hydrogels. ACS APPLIED BIO MATERIALS 2020; 3:2875-2890. [DOI: 10.1021/acsabm.0c00187] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Liu X, Li C, Lv J, Huang F, An Y, Shi L, Ma R. Glucose and H2O2 Dual-Responsive Polymeric Micelles for the Self-Regulated Release of Insulin. ACS APPLIED BIO MATERIALS 2020; 3:1598-1606. [DOI: 10.1021/acsabm.9b01185] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Xiaoyu Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chang Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Juan Lv
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Fan Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yingli An
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Collaborative Innovation Center1 of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Rujiang Ma
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
48
|
Agazzi ML, Herrera SE, Cortez ML, Marmisollé WA, Tagliazucchi M, Azzaroni O. Insulin Delivery from Glucose‐Responsive, Self‐Assembled, Polyamine Nanoparticles: Smart “Sense‐and‐Treat” Nanocarriers Made Easy. Chemistry 2020; 26:2456-2463. [DOI: 10.1002/chem.201905075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Maximiliano L. Agazzi
- Instituto de Investigaciones Fisicoquímicas Teóricas y, Aplicadas Facultad de Ciencias ExactasUniversidad Nacional de La, Plata-CONICET Sucursal 4, Casilla de Correo 16 1900 La Plata Argentina
| | - Santiago E. Herrera
- Instituto de Investigaciones Fisicoquímicas Teóricas y, Aplicadas Facultad de Ciencias ExactasUniversidad Nacional de La, Plata-CONICET Sucursal 4, Casilla de Correo 16 1900 La Plata Argentina
| | - M. Lorena Cortez
- Instituto de Investigaciones Fisicoquímicas Teóricas y, Aplicadas Facultad de Ciencias ExactasUniversidad Nacional de La, Plata-CONICET Sucursal 4, Casilla de Correo 16 1900 La Plata Argentina
| | - Waldemar A. Marmisollé
- Instituto de Investigaciones Fisicoquímicas Teóricas y, Aplicadas Facultad de Ciencias ExactasUniversidad Nacional de La, Plata-CONICET Sucursal 4, Casilla de Correo 16 1900 La Plata Argentina
| | - Mario Tagliazucchi
- Departamento de Química Inorgánica, Analítica y Química FísicaINQUIMAE-CONICETFacultad de Ciencias Exactas y NaturalesCiudad Universitaria Pabellón 2 Buenos Aires C1428EHA Argentina
| | - Omar Azzaroni
- Instituto de Investigaciones Fisicoquímicas Teóricas y, Aplicadas Facultad de Ciencias ExactasUniversidad Nacional de La, Plata-CONICET Sucursal 4, Casilla de Correo 16 1900 La Plata Argentina
| |
Collapse
|
49
|
Wang Y, Fu M, Wang Z, Zhu XX, Guan Y, Zhang Y. A sustained zero-order release carrier for long-acting, peakless basal insulin therapy. J Mater Chem B 2020; 8:1952-1959. [DOI: 10.1039/c9tb02728a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A new drug carrier, which mimics physiologic basal insulin, and secretes and releases insulin at a constant rate, was designed.
Collapse
Affiliation(s)
- Yuanpeng Wang
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology
- Institute of Polymer Chemistry
- College of Chemistry
- Nankai University
- Tianjin 300071
| | - Mian Fu
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology
- Institute of Polymer Chemistry
- College of Chemistry
- Nankai University
- Tianjin 300071
| | - Zuwei Wang
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology
- Institute of Polymer Chemistry
- College of Chemistry
- Nankai University
- Tianjin 300071
| | - X. X. Zhu
- Département de Chimie
- Université de Montréal
- Canada
| | - Ying Guan
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology
- Institute of Polymer Chemistry
- College of Chemistry
- Nankai University
- Tianjin 300071
| | - Yongjun Zhang
- Key Laboratory of Functional Polymer Materials and State Key Laboratory of Medicinal Chemical Biology
- Institute of Polymer Chemistry
- College of Chemistry
- Nankai University
- Tianjin 300071
| |
Collapse
|
50
|
Chen S, Matsumoto H, Moro-oka Y, Tanaka M, Miyahara Y, Suganami T, Matsumoto A. Smart Microneedle Fabricated with Silk Fibroin Combined Semi-interpenetrating Network Hydrogel for Glucose-Responsive Insulin Delivery. ACS Biomater Sci Eng 2019; 5:5781-5789. [DOI: 10.1021/acsbiomaterials.9b00532] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Siyuan Chen
- Kanagawa Institute of Industrial Science and Technology, Kanagawa 213-0012, Japan
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan
| | - Hiroko Matsumoto
- Kanagawa Institute of Industrial Science and Technology, Kanagawa 213-0012, Japan
| | - Yuki Moro-oka
- Kanagawa Institute of Industrial Science and Technology, Kanagawa 213-0012, Japan
| | - Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Yuji Miyahara
- Kanagawa Institute of Industrial Science and Technology, Kanagawa 213-0012, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Akira Matsumoto
- Kanagawa Institute of Industrial Science and Technology, Kanagawa 213-0012, Japan
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan
| |
Collapse
|