1
|
Ji K, Wei X, Kahkoska AR, Zhang J, Zhang Y, Xu J, Wei X, Liu W, Wang Y, Yao Y, Huang X, Mei S, Liu Y, Wang S, Zhao Z, Lu Z, You J, Xu G, Shen Y, Buse JB, Wang J, Gu Z. An orally administered glucose-responsive polymeric complex for high-efficiency and safe delivery of insulin in mice and pigs. NATURE NANOTECHNOLOGY 2024:10.1038/s41565-024-01764-5. [PMID: 39223256 DOI: 10.1038/s41565-024-01764-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 07/19/2024] [Indexed: 09/04/2024]
Abstract
Contrary to current insulin formulations, endogenous insulin has direct access to the portal vein, regulating glucose metabolism in the liver with minimal hypoglycaemia. Here we report the synthesis of an amphiphilic diblock copolymer comprising a glucose-responsive positively charged segment and polycarboxybetaine. The mixing of this polymer with insulin facilitates the formation of worm-like micelles, achieving highly efficient absorption by the gastrointestinal tract and the creation of a glucose-responsive reservoir in the liver. Under hyperglycaemic conditions, the polymer triggers a rapid release of insulin, establishing a portal-to-peripheral insulin gradient-similarly to endogenous insulin-for the safe regulation of blood glucose. This insulin formulation exhibits a dose-dependent blood-glucose-regulating effect in a streptozotocin-induced mouse model of type 1 diabetes and controls the blood glucose at normoglycaemia for one day in non-obese diabetic mice. In addition, the formulation demonstrates a blood-glucose-lowering effect for one day in a pig model of type 1 diabetes without observable hypoglycaemia, showing promise for the safe and effective management of type 1 diabetes.
Collapse
Affiliation(s)
- Kangfan Ji
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xiangqian Wei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Anna R Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Juan Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yang Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jianchang Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xinwei Wei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Wei Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yuejun Yao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xuehui Huang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Shaoqian Mei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yun Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Shiqi Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Zhengjie Zhao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Ziyi Lu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jiahuan You
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Guangzheng Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - John B Buse
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Liu Y, Wang S, Wang Z, Yu J, Wang J, Buse JB, Gu Z. Recent Progress in Glucose-Responsive Insulin. Diabetes 2024; 73:1377-1388. [PMID: 38857114 DOI: 10.2337/dbi23-0028] [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: 02/12/2024] [Accepted: 05/22/2024] [Indexed: 06/12/2024]
Abstract
Insulin replacement therapy is indispensable in the treatment of type 1 and advanced type 2 diabetes. However, insulin's clinical application is challenging due to its narrow therapeutic index. To mitigate acute and chronic risks of glucose excursions, glucose-responsive insulin (GRI) has long been pursued for clinical application. By integrating GRI with glucose-sensitive elements, GRI is capable of releasing or activating insulin in response to plasma or interstitial glucose levels without external monitoring, thereby improving glycemic control and reducing hypoglycemic risk. In this Perspective, we first introduce the history of GRI development and then review major glucose-responsive components that can be leveraged to control insulin delivery. Subsequently, we highlight the recent advances in GRI delivery carriers and insulin analogs. Finally, we provide a look to the future and the challenges of clinical application of GRI. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yun Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shiqi Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zejun Wang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, China
| | - Jicheng Yu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - John B Buse
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Wang M, Zhong H, Li Y, Li J, Zhang X, He F, Wei P, Wang HH, Nie Z. Advances in Bioinspired Artificial System Enabling Biomarker-Driven Therapy. Chemistry 2024; 30:e202401593. [PMID: 38923644 DOI: 10.1002/chem.202401593] [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: 04/23/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 06/28/2024]
Abstract
Bioinspired molecular engineering strategies have emerged as powerful tools that significantly enhance the development of novel therapeutics, improving efficacy, specificity, and safety in disease treatment. Recent advancements have focused on identifying and utilizing disease-associated biomarkers to optimize drug activity and address challenges inherent in traditional therapeutics, such as frequent drug administrations, poor patient adherence, and increased risk of adverse effects. In this review, we provide a comprehensive overview of the latest developments in bioinspired artificial systems (BAS) that use molecular engineering to tailor therapeutic responses to drugs in the presence of disease-specific biomarkers. We examine the transition from open-loop systems, which rely on external cues, to closed-loop feedback systems capable of autonomous self-regulation in response to disease-associated biomarkers. We detail various BAS modalities designed to achieve biomarker-driven therapy, including activatable prodrug molecules, smart drug delivery platforms, autonomous artificial cells, and synthetic receptor-based cell therapies, elucidating their operational principles and practical in vivo applications. Finally, we discuss the current challenges and future perspectives in the advancement of BAS-enabled technology and envision that ongoing advancements toward more programmable and customizable BAS-based therapeutics will significantly enhance precision medicine.
Collapse
Affiliation(s)
- Meixia Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Huan Zhong
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Yangbing Li
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Juan Li
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Xinxin Zhang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Fang He
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Ping Wei
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Zhou Nie
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| |
Collapse
|
4
|
Wang Y, Shi J, Xin M, Kahkoska AR, Wang J, Gu Z. Cell-drug conjugates. Nat Biomed Eng 2024:10.1038/s41551-024-01230-6. [PMID: 38951139 DOI: 10.1038/s41551-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/01/2024] [Indexed: 07/03/2024]
Abstract
By combining living cells with therapeutics, cell-drug conjugates can potentiate the functions of both components, particularly for applications in drug delivery and therapy. The conjugates can be designed to persist in the bloodstream, undergo chemotaxis, evade surveillance by the immune system, proliferate, or maintain or transform their cellular phenotypes. In this Review, we discuss strategies for the design of cell-drug conjugates with specific functions, the techniques for their preparation, and their applications in the treatment of cancers, autoimmune diseases and other pathologies. We also discuss the translational challenges and opportunities of this class of drug-delivery systems and therapeutics.
Collapse
Affiliation(s)
- Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jiaqi Shi
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Minhang Xin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Anna R Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
5
|
Varas N, Grabowski R, Jarosinski MA, Tai N, Herzog RI, Ismail-Beigi F, Yang Y, Cherrington AD, Weiss MA. Ultra-stable insulin-glucagon fusion protein exploits an endogenous hepatic switch to mitigate hypoglycemic risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.594997. [PMID: 38826486 PMCID: PMC11142066 DOI: 10.1101/2024.05.20.594997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The risk of hypoglycemia and its serious medical sequelae restrict insulin replacement therapy for diabetes mellitus. Such adverse clinical impact has motivated development of diverse glucose-responsive technologies, including algorithm-controlled insulin pumps linked to continuous glucose monitors ("closed-loop systems") and glucose-sensing ("smart") insulins. These technologies seek to optimize glycemic control while minimizing hypoglycemic risk. Here, we describe an alternative approach that exploits an endogenous glucose-dependent switch in hepatic physiology: preferential insulin signaling (under hyperglycemic conditions) versus preferential counter-regulatory glucagon signaling (during hypoglycemia). Motivated by prior reports of glucagon-insulin co-infusion, we designed and tested an ultra-stable glucagon-insulin fusion protein whose relative hormonal activities were calibrated by respective modifications; physical stability was concurrently augmented to facilitate formulation, enhance shelf life and expand access. An N-terminal glucagon moiety was stabilized by an α-helix-compatible Lys 13 -Glu 17 lactam bridge; A C-terminal insulin moiety was stabilized as a single chain with foreshortened C domain. Studies in vitro demonstrated (a) resistance to fibrillation on prolonged agitation at 37 °C and (b) dual hormonal signaling activities with appropriate balance. Glucodynamic responses were monitored in rats relative to control fusion proteins lacking one or the other hormonal activity, and continuous intravenous infusion emulated basal subcutaneous therapy. Whereas efficacy in mitigating hyperglycemia was unaffected by the glucagon moiety, the fusion protein enhanced endogenous glucose production under hypoglycemic conditions. Together, these findings provide proof of principle toward a basal glucose-responsive insulin biotechnology of striking simplicity. The fusion protein's augmented stability promises to circumvent the costly cold chain presently constraining global insulin access. Significance Statement The therapeutic goal of insulin replacement therapy in diabetes is normalization of blood-glucose concentration, which prevents or delays long-term complications. A critical barrier is posed by recurrent hypoglycemic events that results in short- and long-term morbidities. An innovative approach envisions co-injection of glucagon (a counter-regulatory hormone) to exploit a glycemia-dependent hepatic switch in relative hormone responsiveness. To provide an enabling technology, we describe an ultra-stable fusion protein containing insulin- and glucagon moieties. Proof of principle was obtained in rats. A single-chain insulin moiety provides glycemic control whereas a lactam-stabilized glucagon extension mitigates hypoglycemia. This dual-hormone fusion protein promises to provide a basal formulation with reduced risk of hypoglycemia. Resistance to fibrillation may circumvent the cold chain required for global access.
Collapse
|
6
|
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
|
7
|
Yang S, Yang JF, Gong X, Weiss MA, Strano MS. Rational Design and Efficacy of Glucose-Responsive Insulin Therapeutics and Insulin Delivery Systems by Computation Using Connected Human and Rodent Models. Adv Healthc Mater 2023; 12:e2300587. [PMID: 37319398 PMCID: PMC10592437 DOI: 10.1002/adhm.202300587] [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: 02/22/2023] [Revised: 06/02/2023] [Indexed: 06/17/2023]
Abstract
Glucose-responsive insulins (GRIs) use plasma glucose levels in a diabetic patient to activate a specifically designed insulin analogue to a more potent state in real time. Alternatively, some GRI concepts use glucose-mediated release or injection of insulin into the bloodstream. GRIs hold promise to exhibit much improved pharmacological control of the plasma glucose concentration, particularly for the problem of therapeutically induced hypoglycemia. Several innovative GRI schemes are introduced into the literature, but there remains a dearth of quantitative analysis to aid the development and optimization of these constructs into effective therapeutics. This work evaluates several classes of GRIs that are proposed using a pharmacokinetic model as previously described, PAMERAH, simulating the glucoregulatory system of humans and rodents. GRI concepts are grouped into three mechanistic classes: 1) intrinsic GRIs, 2) glucose-responsive particles, and 3) glucose-responsive devices. Each class is analyzed for optimal designs that maintain glucose levels within the euglycemic range. These derived GRI parameter spaces are then compared between rodents and humans, providing the differences in clinical translation success for each candidate. This work demonstrates a computational framework to evaluate the potential clinical translatability of existing glucose-responsive systems, providing a useful approach for future GRI development.
Collapse
Affiliation(s)
- Sungyun Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jing Fan Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Xun Gong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University of Medicine, Indianapolis, IN, 46202, USA
| | - Michael S Strano
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
8
|
Ubba V, Joseph S, Awe O, Jones D, Dsilva MK, Feng M, Wang J, Fu X, Akbar RJ, Bodnar BH, Hu W, Wang H, Yang X, Yang L, Yang P, Ahima R, Divall S, Wu S. Neuronal AR Regulates Glucose Homeostasis and Energy Expenditure in Lean Female Mice With Androgen Excess. Endocrinology 2023; 164:bqad141. [PMID: 37738624 DOI: 10.1210/endocr/bqad141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023]
Abstract
Hyperandrogenemia and polycystic ovary syndrome are a result of the imbalance of androgen levels in females. Androgen receptor (Ar) mediates the effect of androgen, and this study examines how neuronal Ar in the central nervous system mediates metabolism under normal and increased androgen conditions in female mice. The neuron-specific ARKO mouse (SynARKO) was created from female (Ar fl/wt; synapsin promoter driven Cre) and male (Ar fl/y) mice. A glucose tolerance test revealed impaired glucose tolerance that was partially alleviated in the SynARKO-dihydrotestosterone (DHT) mice compared with Con-DHT mice after 4 months of DHT treatment. Heat production and food intake was higher in Con-DHT mice than in Con-veh mice; these effects were not altered between SynARKO-veh and SynARKO-DHT mice, indicating that excess androgens may partially alter calorie intake and energy expenditure in females via the neuronal Ar. The pAkt/Akt activity was higher in the hypothalamus in Con-DHT mice than in Con-veh mice, and this effect was attenuated in SynARKO-DHT mice. Western blot studies show that markers of inflammation and microglia activation, such as NF-kB p-65 and IBA1, increased in the hypothalamus of Con-DHT mice compared with Con-veh. These studies suggest that neuronal Ar mediates the metabolic impacts of androgen excess in females.
Collapse
Affiliation(s)
- Vaibhave Ubba
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Serene Joseph
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Olubusayo Awe
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Dustin Jones
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Milan K Dsilva
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Mingxiao Feng
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21087, USA
| | - Junjiang Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21087, USA
| | - Xiaomin Fu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21087, USA
| | - Razeen J Akbar
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Brittany H Bodnar
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Wenhui Hu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Ling Yang
- Department of Medical Genetics & Molecular Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rexford Ahima
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sara Divall
- Department of Pediatrics, University of Washington, Seattle's Children's Hospital, Seattle, WA 98145-5005, USA
| | - Sheng Wu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21087, USA
| |
Collapse
|
9
|
Ji K, Yao Y, Wei X, Liu W, Zhang J, Liu Y, Zhang Y, Wang J, Gu Z. Material design for oral insulin delivery. MED-X 2023; 1:7. [PMID: 37485249 PMCID: PMC10357414 DOI: 10.1007/s44258-023-00006-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 07/25/2023]
Abstract
Frequent insulin injections remain the primary method for controlling the blood glucose level of individuals with diabetes mellitus but are associated with low compliance. Accordingly, oral administration has been identified as a highly desirable alternative due to its non-invasive nature. However, the harsh gastrointestinal environment and physical intestinal barriers pose significant challenges to achieving optimal pharmacological bioavailability of insulin. As a result, researchers have developed a range of materials to improve the efficiency of oral insulin delivery over the past few decades. In this review, we summarize the latest advances in material design that aim to enhance insulin protection, permeability, and glucose-responsive release. We also explore the opportunities and challenges of using these materials for oral insulin delivery.
Collapse
Affiliation(s)
- Kangfan Ji
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Yuejun Yao
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Xinwei Wei
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Wei Liu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Juan Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Yun Liu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Yang Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Jinqiang Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
- Department of General Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016 China
- Zhejiang Laboratory of Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121 China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| |
Collapse
|
10
|
Zheng W, Quan B, Gao G, Zhang P, Huang L. Combination of Circulating Cell-Free DNA and Positron Emission Tomography to Distinguish Non-Small Cell Lung Cancer from Tuberculosis. Lab Med 2023; 54:130-141. [PMID: 36106407 DOI: 10.1093/labmed/lmac085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Non-small cell lung cancer (NSCLC) holds high metabolic tumor burden and circulating cell-free DNA (cfDNA) levels, and the relationship between metabolic tumor burden and cfDNA in NSCLC and the underlying mechanism of their interaction therein remain poorly characterized. Our aim was to evaluate the clinical value of cfDNA and metabolic tumor burden by positron emission tomography-computed tomography (PET/CT) for NSCLC differential diagnosis from tuberculosis in patients with solitary pulmonary nodules. METHODS Metabolic tumor burden values in humans (subjects with NSCLC, subjects with tuberculosis, and healthy control subjects) and relevant mouse models were detected by preoperative 18F-fluorodeoxyglucose PET (18F-FDG PET/CT) and [3H]-2-deoxy-DG uptake, respectively. The cfDNA levels were detected by quantifying serum cfDNA fragments from the ALU (115 bp) gene using reverse transcription-polymerase chain reaction. RNA sequence was performed to determine the underlying target genes and knocked down or inhibited the target genes in vivo and in vitro to determine the mechanism therein. RESULTS Metabolic tumor burden correlated with serum cfDNA levels in NSCLC subjects but not in tuberculosis subjects or healthy controls. Mouse models showed a similar phenomenon. In addition, the RNA sequence showed that glucose transporter 1 (GLU1), factor-related apoptosis ligand (FasL), caspase 8, and caspase 3 were significantly increased in NSCLC mouse tumors compared with those in tuberculosis mouse masses. Inhibiting the metabolic tumor burden by blocking or knocking down GLU1 markedly reduced the expression of FasL, the phosphorylation of caspase 8/caspase 3, and serum cfDNA levels/apoptosis percentage in vivo and in vitro. Furthermore, the use of a combination of cfDNA and metabolic tumor burden allowed better ability to distinguish NSCLC subjects from those with tuberculosis or healthy controls than either method used alone. CONCLUSION Metabolic tumor burden promotes the formation of circulating cfDNA through GLU1-mediated apoptosis in NSCLC, and the combination of cfDNA and metabolic tumor burden could be valuable for distinguishing NSCLC from tuberculosis.
Collapse
Affiliation(s)
- Wenqiang Zheng
- Department of Nuclear Medicine, First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Bin Quan
- Department of Infectious Diseases, First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Guangjian Gao
- Department of Nuclear Medicine, First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Puhong Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Lizhu Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
11
|
Bolli GB, Cheng AYY, Owens DR. Insulin: evolution of insulin formulations and their application in clinical practice over 100 years. Acta Diabetol 2022; 59:1129-1144. [PMID: 35854185 PMCID: PMC9296014 DOI: 10.1007/s00592-022-01938-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 07/01/2022] [Indexed: 11/22/2022]
Abstract
The first preparation of insulin extracted from a pancreas and made suitable for use in humans after purification was achieved 100 years ago in Toronto, an epoch-making achievement, which has ultimately provided a life-giving treatment for millions of people worldwide. The earliest animal-derived formulations were short-acting and contained many impurities that caused adverse reactions, thereby limiting their therapeutic potential. However, since then, insulin production and purification improved with enhanced technologies, along with a full understanding of the insulin molecule structure. The availability of radio-immunoassays contributed to the unravelling of the physiology of glucose homeostasis, ultimately leading to the adoption of rational models of insulin replacement. The introduction of recombinant DNA technologies has since resulted in the era of both rapid- and long-acting human insulin analogues administered via the subcutaneous route which better mimic the physiology of insulin secretion, leading to the modern basal-bolus regimen. These advances, in combination with improved education and technologies for glucose monitoring, enable people with diabetes to better meet individual glycaemic goals with a lower risk of hypoglycaemia. While the prevalence of diabetes continues to rise globally, it is important to recognise the scientific endeavour that has led to insulin remaining the cornerstone of diabetes management, on the centenary of its first successful use in humans.
Collapse
|
12
|
Molecular basis for inhibiting human glucose transporters by exofacial inhibitors. Nat Commun 2022; 13:2632. [PMID: 35552392 PMCID: PMC9098912 DOI: 10.1038/s41467-022-30326-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 04/26/2022] [Indexed: 12/27/2022] Open
Abstract
Human glucose transporters (GLUTs) are responsible for cellular uptake of hexoses. Elevated expression of GLUTs, particularly GLUT1 and GLUT3, is required to fuel the hyperproliferation of cancer cells, making GLUT inhibitors potential anticancer therapeutics. Meanwhile, GLUT inhibitor-conjugated insulin is being explored to mitigate the hypoglycemia side effect of insulin therapy in type 1 diabetes. Reasoning that exofacial inhibitors of GLUT1/3 may be favored for therapeutic applications, we report here the engineering of a GLUT3 variant, designated GLUT3exo, that can be probed for screening and validating exofacial inhibitors. We identify an exofacial GLUT3 inhibitor SA47 and elucidate its mode of action by a 2.3 Å resolution crystal structure of SA47-bound GLUT3. Our studies serve as a framework for the discovery of GLUTs exofacial inhibitors for therapeutic development. Human glucose transporters (GLUTs), particularly GLUT1 and GLUT3, are potential anticancer therapy targets. Here, Nan Wang et al. use an engineered GLUT 3 variant to identify an exofacial GLUT3 inhibitor, SA47, and elucidate the drug’s inhibitory mechanism.
Collapse
|
13
|
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: 3.0] [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
|
14
|
Zhang W, Zhang W, Li C, Zhang J, Qin L, Lai Y. Recent Advances of Microneedles and Their Application in Disease Treatment. Int J Mol Sci 2022; 23:2401. [PMID: 35269545 PMCID: PMC8909978 DOI: 10.3390/ijms23052401] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/21/2022] Open
Abstract
For decades, scientists have been doing a lot of research and exploration to find effective long-term analgesic and/or disease-modifying treatments. Microneedles (MNs) are a simple, effective, and painless transdermal drug delivery technology that has emerged in recent years, and exhibits great promise for realizing intelligent drug delivery. With the development of materials science and fabrication technology, the MN transdermal drug delivery technology has been applied and popularized in more and more fields, including chronic illnesses such as arthritis or diabetes, cancer, dermatocosmetology, family planning, and epidemic disease prevention, and has made fruitful achievements. This paper mainly reviews the latest research status of MNs and their fabrication methodology, and summarizes the application of MNs in the treatment of various diseases, as well as the potential to use nanotechnology to develop more intelligent MNs-based drug delivery systems.
Collapse
Affiliation(s)
- Wenjing Zhang
- Center for Translational Medicine Research and Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.Z.); (W.Z.); (C.L.); (J.Z.); (L.Q.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Zhang
- Center for Translational Medicine Research and Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.Z.); (W.Z.); (C.L.); (J.Z.); (L.Q.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Cairong Li
- Center for Translational Medicine Research and Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.Z.); (W.Z.); (C.L.); (J.Z.); (L.Q.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianhua Zhang
- Center for Translational Medicine Research and Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.Z.); (W.Z.); (C.L.); (J.Z.); (L.Q.)
| | - Ling Qin
- Center for Translational Medicine Research and Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.Z.); (W.Z.); (C.L.); (J.Z.); (L.Q.)
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, China
- CAS-HK Joint Lab of Biomaterials, Shenzhen 518055, China
| | - Yuxiao Lai
- Center for Translational Medicine Research and Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.Z.); (W.Z.); (C.L.); (J.Z.); (L.Q.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS-HK Joint Lab of Biomaterials, Shenzhen 518055, China
- Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
15
|
Battistella C, Liang Y, Gianneschi NC. Innovations in Disease State Responsive Soft Materials for Targeting Extracellular Stimuli Associated with Cancer, Cardiovascular Disease, Diabetes, and Beyond. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007504. [PMID: 34145625 PMCID: PMC9836048 DOI: 10.1002/adma.202007504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/20/2021] [Indexed: 05/10/2023]
Abstract
Recent advances in polymer chemistry, materials sciences, and biotechnology have allowed the preclinical development of sophisticated programmable nanomedicines and materials that are able to precisely respond to specific disease-associated triggers and microenvironments. These stimuli, endogenous to the targeted diseases, include pH, redox-state, small molecules, and protein upregulation. Herein, recent advances and innovative approaches in programmable soft materials capable of sensing the aforementioned disease-associated stimuli and responding via a range of dynamic processes including morphological and size transitions, changes in mobility and retention, as well as disassembly are described. In this field generally, the majority of ongoing and past research effort has focused on oncology. Given this interest, examples of the latest innovative approaches to chemo- and immunotherapy treatment strategies for cancer are presented. Moreover, as the field broadens its attention, applications of programmable materials in other diseases are highlighted, with a special focus on cardiovascular disease and diabetes mellitus, where limited attention is paid by the field, but where many promising avenues exist with high potential impact.
Collapse
Affiliation(s)
- Claudia Battistella
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Department of Biomedical Engineering, Department of Pharmacology, Northwestern University, Evanston, IL, 60208, USA
| | - Yifei Liang
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Nathan C Gianneschi
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Department of Biomedical Engineering, Department of Pharmacology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
16
|
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: 4.7] [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
|
17
|
Kurtzhals P, Nishimura E, Haahr H, Høeg-Jensen T, Johansson E, Madsen P, Sturis J, Kjeldsen T. Commemorating insulin's centennial: engineering insulin pharmacology towards physiology. Trends Pharmacol Sci 2021; 42:620-639. [PMID: 34148677 DOI: 10.1016/j.tips.2021.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 01/14/2023]
Abstract
The life-saving discovery of insulin in Toronto in 1921 is one of the most impactful achievements in medical history, at the time being hailed as a miracle treatment for diabetes. The insulin molecule itself, however, is poorly amenable as a pharmacological intervention, and the formidable challenge of optimizing insulin therapy has been ongoing for a century. We review early academic insights into insulin structure and its relation to self-association and receptor binding, as well as recombinant biotechnology, which have all been seminal for drug design. Recent developments have focused on combining genetic and chemical engineering with pharmaceutical optimization to generate ultra-rapid and ultra-long-acting, tissue-selective, or orally delivered insulin analogs. We further discuss these developments and propose that future scientific efforts in molecular engineering include realizing the dream of glucose-responsive insulin delivery.
Collapse
Affiliation(s)
- Peter Kurtzhals
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark.
| | - Erica Nishimura
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Hanne Haahr
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Thomas Høeg-Jensen
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Eva Johansson
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Peter Madsen
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Jeppe Sturis
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Thomas Kjeldsen
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| |
Collapse
|
18
|
Szunerits S, Melinte S, Barras A, Pagneux Q, Voronova A, Abderrahmani A, Boukherroub R. The impact of chemical engineering and technological advances on managing diabetes: present and future concepts. Chem Soc Rev 2021; 50:2102-2146. [PMID: 33325917 DOI: 10.1039/c9cs00886a] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Monitoring blood glucose levels for diabetic patients is critical to achieve tight glycaemic control. As none of the current antidiabetic treatments restore lost functional β-cell mass in diabetic patients, insulin injections and the use of insulin pumps are most widely used in the management of glycaemia. The use of advanced and intelligent chemical engineering, together with the incorporation of micro- and nanotechnological-based processes have lately revolutionized diabetic management. The start of this concept goes back to 1974 with the description of an electrode that repeatedly measures the level of blood glucose and triggers insulin release from an infusion pump to enter the blood stream from a small reservoir upon need. Next to the insulin pumps, other drug delivery routes, including nasal, transdermal and buccal, are currently investigated. These processes necessitate competences from chemists, engineers-alike and innovative views of pharmacologists and diabetologists. Engineered micro and nanostructures hold a unique potential when it comes to drug delivery applications required for the treatment of diabetic patients. As the technical aspects of chemistry, biology and informatics on medicine are expanding fast, time has come to step back and to evaluate the impact of technology-driven chemistry on diabetics and how the bridges from research laboratories to market products are established. In this review, the large variety of therapeutic approaches proposed in the last five years for diabetic patients are discussed in an applied context. A survey of the state of the art of closed-loop insulin delivery strategies in response to blood glucose level fluctuation is provided together with insights into the emerging key technologies for diagnosis and drug development. Chemical engineering strategies centered on preserving and regenerating functional pancreatic β-cell mass are evoked in addition as they represent a permanent solution for diabetic patients.
Collapse
Affiliation(s)
- Sabine Szunerits
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN, F-59000 Lille, France.
| | - Sorin Melinte
- Institute of Information and Communication Technologies, Electronics and Applied Mathematics, Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Alexandre Barras
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN, F-59000 Lille, France.
| | - Quentin Pagneux
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN, F-59000 Lille, France.
| | - Anna Voronova
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN, F-59000 Lille, France.
| | - Amar Abderrahmani
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN, F-59000 Lille, France.
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN, F-59000 Lille, France.
| |
Collapse
|
19
|
A step towards glucose control with a novel nanomagnetic-insulin for diabetes care. Int J Pharm 2021; 601:120587. [PMID: 33845153 DOI: 10.1016/j.ijpharm.2021.120587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/28/2021] [Accepted: 04/04/2021] [Indexed: 01/06/2023]
Abstract
Massive efforts have been devoted to insulin delivery for diabetes care. Achieving a long-term tight-regulated blood glucose level with a low risk of hypoglycemia remains a great challenge. In this study we propose a novel strategy to efficiently regulate insulin action after insulin is injected or released into patient body aiming to achieve better glycemic control, which is achieved by the administration of insulin-conjugated magnetic nanoparticles (MNPs-Ins). We show that the locomotion of MNPs-Ins can be controlled to reach a target site on an in vitro microfluidic platform, which may open a way to modulate the physiological effect of insulin in a remote-control manner. Most importantly, the in vivo blood glucose regulation of the MNPs-Ins was performed on diabetic mice to understand the glycemic control performance. The results showed that the MNPs-Ins can achieve a better glycemic control with longer effective drug duration while not causing hypoglycemia and a magnetic-modulated hypoglycemic dynamics. Moreover, the in vivo histochemistry experiments confirmed the good biocompatibility of MNPs-Ins. Along with our on-going research on the possibility of the recycle and reuse of the MNPs-Ins, the finding presented in this paper may manifest a fascinating potential in insulin delivery in the near future.
Collapse
|
20
|
He M, Yu P, Hu Y, Zhang J, He M, Nie C, Chu X. Erythrocyte-Membrane-Enveloped Biomineralized Metal-Organic Framework Nanoparticles Enable Intravenous Glucose-Responsive Insulin Delivery. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19648-19659. [PMID: 33890785 DOI: 10.1021/acsami.1c01943] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A "closed-loop" insulin delivery system that can mimic the dynamic and glucose-responsive insulin secretion as islet β-cells is desirable for the therapy of type 1 and advanced type 2 diabetes mellitus (T1DM and T2DM). Herein, we introduced a kind of "core-shell"-structured glucose-responsive nanoplatform to achieve intravenous "smart" insulin delivery. A finely controlled one-pot biomimetic mineralization method was utilized to coencapsulate insulin, glucose oxidase (GOx), and catalase (CAT) into the ZIF-8 nanoparticles (NPs) to construct the "inner core", where an efficient enzyme cascade system (GOx/CAT group) served as an optimized glucose-responsive module that could rapidly catalyze glucose to yield gluconic acid to lower the local pH and effectively consume the harmful byproduct hydrogen peroxide (H2O2), inducing the collapse of pH-sensitive ZIF-8 NPs to release insulin. The erythrocyte membrane, a sort of natural biological derived lipid bilayer membrane which has intrinsic biocompatibility, was enveloped onto the surface of the "inner core" as the "outer shell" to protect them from elimination by the immune system, thus making the NPs intravenously injectable and could stably maintain a long-term existence in blood circulation. The in vitro and in vivo results indicate that our well-designed nanoplatform possesses an excellent glucose-responsive property and can maintain the blood glucose levels of the streptozocin (STZ)-induced type 1 diabetic mice at the normoglycemic state for up to 24 h after being intravenously administrated, confirming an intravenous insulin delivery strategy to overcome the deficits of conventional daily multiple subcutaneous insulin administration and offering a potential candidate for long-term T1DM treatment.
Collapse
Affiliation(s)
- Mengyun He
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Pei Yu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Yanlei Hu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Juan Zhang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Manman He
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Cunpeng Nie
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Xia Chu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
21
|
Jarosinski MA, Dhayalan B, Rege N, Chatterjee D, Weiss MA. 'Smart' insulin-delivery technologies and intrinsic glucose-responsive insulin analogues. Diabetologia 2021; 64:1016-1029. [PMID: 33710398 PMCID: PMC8158166 DOI: 10.1007/s00125-021-05422-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Insulin replacement therapy for diabetes mellitus seeks to minimise excursions in blood glucose concentration above or below the therapeutic range (hyper- or hypoglycaemia). To mitigate acute and chronic risks of such excursions, glucose-responsive insulin-delivery technologies have long been sought for clinical application in type 1 and long-standing type 2 diabetes mellitus. Such 'smart' systems or insulin analogues seek to provide hormonal activity proportional to blood glucose levels without external monitoring. This review highlights three broad strategies to co-optimise mean glycaemic control and time in range: (1) coupling of continuous glucose monitoring (CGM) to delivery devices (algorithm-based 'closed-loop' systems); (2) glucose-responsive polymer encapsulation of insulin; and (3) mechanism-based hormone modifications. Innovations span control algorithms for CGM-based insulin-delivery systems, glucose-responsive polymer matrices, bio-inspired design based on insulin's conformational switch mechanism upon insulin receptor engagement, and glucose-responsive modifications of new insulin analogues. In each case, innovations in insulin chemistry and formulation may enhance clinical outcomes. Prospects are discussed for intrinsic glucose-responsive insulin analogues containing a reversible switch (regulating bioavailability or conformation) that can be activated by glucose at high concentrations.
Collapse
Affiliation(s)
- Mark A Jarosinski
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nischay Rege
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Deepak Chatterjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Chemistry, Indiana University, Bloomington, IN, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
22
|
Zhang Y, Xie Y, Hao Z, Zhou P, Wang P, Fang S, Li L, Xu S, Xia Y. Umbilical Mesenchymal Stem Cell-Derived Exosome-Encapsulated Hydrogels Accelerate Bone Repair by Enhancing Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:18472-18487. [PMID: 33856781 DOI: 10.1021/acsami.0c22671] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Repair of large bone defects represents a major challenge for orthopedic surgeons. The newly formed microvessels inside grafts play a crucial role in successful bone tissue engineering. Previously, an active role for mesenchymal stem cell (MSC)-derived exosomes in blood vessel development and progression was suggested in the repair of multiple tissues. However, the reports on the application of MSC-derived exosomes in the repair of large bone defects are sparse. In this study, we encapsulated umbilical MSC-derived exosomes (uMSCEXOs) in hyaluronic acid hydrogel (HA-Gel) and combined them with customized nanohydroxyapatite/poly-ε-caprolactone (nHP) scaffolds to repair cranial defects in rats. Imaging and histological evaluation indicated that the uMSCEXOs/Gel/nHP composites markedly enhanced bone regeneration in vivo, and the uMSCEXOs might play a key role in this process. Moreover, the in vitro results demonstrated that uMSCEXOs promoted the proliferation, migration, and angiogenic differentiation of endothelial progenitor cells (EPCs) but did not significantly affect the osteogenic differentiation of BMSCs. Importantly, mechanistic studies revealed that exosomal miR-21 was the potential intercellular messenger that promoted angiogenesis by upregulating the NOTCH1/DLL4 pathway. In conclusion, our findings exhibit a promising exosome-based strategy in repairing large bone defects through enhanced angiogenesis, which potentially regulated by the miR-21/NOTCH1/DLL4 signaling axis.
Collapse
Affiliation(s)
- Yuntong Zhang
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yang Xie
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zichen Hao
- Department of Orthopaedics, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
| | - Panyu Zhou
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Panfeng Wang
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shuo Fang
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Lu Li
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shuogui Xu
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Xia
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
23
|
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
|
24
|
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: 21] [Impact Index Per Article: 7.0] [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
|
25
|
Maikawa CL, d'Aquino AI, Lal RA, Buckingham BA, Appel EA. Engineering biopharmaceutical formulations to improve diabetes management. Sci Transl Med 2021; 13:eabd6726. [PMID: 33504649 PMCID: PMC8004356 DOI: 10.1126/scitranslmed.abd6726] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
Insulin was first isolated almost a century ago, yet commercial formulations of insulin and its analogs for hormone replacement therapy still fall short of appropriately mimicking endogenous glycemic control. Moreover, the controlled delivery of complementary hormones (such as amylin or glucagon) is complicated by instability of the pharmacologic agents and complexity of maintaining multiple infusions. In this review, we highlight the advantages and limitations of recent advances in drug formulation that improve protein stability and pharmacokinetics, prolong drug delivery, or enable alternative dosage forms for the management of diabetes. With controlled delivery, these formulations could improve closed-loop glycemic control.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Andrea I d'Aquino
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Rayhan A Lal
- Department of Medicine (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Bruce A Buckingham
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
26
|
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: 24] [Impact Index Per Article: 8.0] [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
|
27
|
Decuzzi P, Peer D, Di Mascolo D, Palange AL, Manghnani PN, Moghimi SM, Farhangrazi ZS, Howard KA, Rosenblum D, Liang T, Chen Z, Wang Z, Zhu JJ, Gu Z, Korin N, Letourneur D, Chauvierre C, van der Meel R, Kiessling F, Lammers T. Roadmap on nanomedicine. NANOTECHNOLOGY 2021; 32:012001. [PMID: 33043901 PMCID: PMC7612035 DOI: 10.1088/1361-6528/abaadb] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Since the launch of the Alliance for Nanotechnology in Cancer by the National Cancer Institute in late 2004, several similar initiatives have been promoted all over the globe with the intention of advancing the diagnosis, treatment and prevention of cancer in the wake of nanoscience and nanotechnology. All this has encouraged scientists with diverse backgrounds to team up with one another, learn from each other, and generate new knowledge at the interface between engineering, physics, chemistry and biomedical sciences. Importantly, this new knowledge has been wisely channeled towards the development of novel diagnostic, imaging and therapeutic nanosystems, many of which are currently at different stages of clinical development. This roadmap collects eight brief articles elaborating on the interaction of nanomedicines with human biology; the biomedical and clinical applications of nanomedicines; and the importance of patient stratification in the development of future nanomedicines. The first article reports on the role of geometry and mechanical properties in nanomedicine rational design; the second articulates on the interaction of nanomedicines with cells of the immune system; and the third deals with exploiting endogenous molecules, such as albumin, to carry therapeutic agents. The second group of articles highlights the successful application of nanomedicines in the treatment of cancer with the optimal delivery of nucleic acids, diabetes with the sustained and controlled release of insulin, stroke by using thrombolytic particles, and atherosclerosis with the development of targeted nanoparticles. Finally, the last contribution comments on how nanomedicine and theranostics could play a pivotal role in the development of personalized medicines. As this roadmap cannot cover the massive extent of development of nanomedicine over the past 15 years, only a few major achievements are highlighted as the field progressively matures from the initial hype to the consolidation phase.
Collapse
Affiliation(s)
- Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
- Corresponding authors: and
| | - Dan Peer
- Laboratory of Precision NanoMedicine, School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering
- Center for Nanoscience and Nanotechnology
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 6997801, Israel
- Corresponding authors: and
| | - Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - Anna Lisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - Purnima Naresh Manghnani
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - S. Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | | | - Kenneth A. Howard
- Interdisciplinary Nanoscience Center, Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - Daniel Rosenblum
- Laboratory of Precision NanoMedicine, School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering
- Center for Nanoscience and Nanotechnology
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Tingxizi Liang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhaowei Chen
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Netanel Korin
- Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Didier Letourneur
- Université de Paris, Université Paris 13, INSERM 1148, LVTS, Hôpital Bichat, F-75018 Paris, France
| | - Cédric Chauvierre
- Université de Paris, Université Paris 13, INSERM 1148, LVTS, Hôpital Bichat, F-75018 Paris, France
| | - Roy van der Meel
- Laboratory of Chemical Biology, Dept. of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
- Dept. of Targeted Therapeutics, University of Twente, Enschede, The Netherlands
- Dept. of Pharmaceutics, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
28
|
Yu J, Wang J, Zhang Y, Chen G, Mao W, Ye Y, Kahkoska AR, Buse JB, Langer R, Gu Z. Glucose-responsive insulin patch for the regulation of blood glucose in mice and minipigs. Nat Biomed Eng 2020; 4:499-506. [PMID: 32015407 PMCID: PMC7231631 DOI: 10.1038/s41551-019-0508-y] [Citation(s) in RCA: 310] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022]
Abstract
Glucose-responsive insulin delivery systems that mimic pancreatic endocrine function could enhance health and improve quality of life for people with type 1 and type 2 diabetes with reduced β-cell function. However, insulin delivery systems with rapid in vivo glucose-responsive behaviour typically have limited insulin-loading capacities and cannot be manufactured easily. Here, we show that a single removable transdermal patch, bearing microneedles loaded with insulin and a non-degradable glucose-responsive polymeric matrix, and fabricated via in situ photopolymerization, regulated blood glucose in insulin-deficient diabetic mice and minipigs (for minipigs >25 kg, glucose regulation lasted >20 h with patches of ~5 cm2). Under hyperglycaemic conditions, phenylboronic acid units within the polymeric matrix reversibly form glucose-boronate complexes that-owing to their increased negative charge-induce the swelling of the polymeric matrix and weaken the electrostatic interactions between the negatively charged insulin and polymers, promoting the rapid release of insulin. This proof-of-concept demonstration may aid the development of other translational stimuli-responsive microneedle patches for drug delivery.
Collapse
Affiliation(s)
- Jicheng Yu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
- Zenomics Inc., Los Angeles, CA, USA
| | - Jinqiang Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuqi Zhang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
- Zenomics Inc., Los Angeles, CA, USA
| | - Guojun Chen
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
- Zenomics Inc., Los Angeles, CA, USA
| | - Anna R Kahkoska
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - John B Buse
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA
- Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
29
|
|
30
|
Zhang C, Hong S, Liu MD, Yu WY, Zhang MK, Zhang L, Zeng X, Zhang XZ. pH-sensitive MOF integrated with glucose oxidase for glucose-responsive insulin delivery. J Control Release 2020; 320:159-167. [DOI: 10.1016/j.jconrel.2020.01.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
|
31
|
Wang J, Wang Z, Yu J, Kahkoska AR, Buse JB, Gu Z. Glucose-Responsive Insulin and Delivery Systems: Innovation and Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1902004. [PMID: 31423670 PMCID: PMC7141789 DOI: 10.1002/adma.201902004] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/09/2019] [Indexed: 05/18/2023]
Abstract
Type 1 and advanced type 2 diabetes treatment involves daily injections or continuous infusion of exogenous insulin aimed at regulating blood glucose levels in the normoglycemic range. However, current options for insulin therapy are limited by the risk of hypoglycemia and are associated with suboptimal glycemic control outcomes. Therefore, a range of glucose-responsive components that can undergo changes in conformation or show alterations in intermolecular binding capability in response to glucose stimulation has been studied for ultimate integration into closed-loop insulin delivery or "smart insulin" systems. Here, an overview of the evolution and recent progress in the development of molecular approaches for glucose-responsive insulin delivery systems, a rapidly growing subfield of precision medicine, is presented. Three central glucose-responsive moieties, including glucose oxidase, phenylboronic acid, and glucose-binding molecules are examined in detail. Future opportunities and challenges regarding translation are also discussed.
Collapse
Affiliation(s)
- Jinqiang Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | | | - 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
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
- Zenomics Inc., Durham, NC 27709, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
32
|
Rudra A, Li J, Shakur R, Bhagchandani S, Langer R. Trends in Therapeutic Conjugates: Bench to Clinic. Bioconjug Chem 2020; 31:462-473. [PMID: 31990184 DOI: 10.1021/acs.bioconjchem.9b00828] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, therapeutic conjugates have attracted considerable attention as a new class of drug due to their unique pharmacological properties, especially from the pharmaceutical community. Their molecular structure tunability, improved targeting specificity, and therapeutic efficacy have been demonstrated in a wide range of research and clinical applications. In this topical review, we summarize selected recent advances in bioconjugation strategies for the development of therapeutic conjugates, their emerging application in clinical settings, as well as perspectives on the direction of future research.
Collapse
Affiliation(s)
- Arnab Rudra
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Junwei Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Rameen Shakur
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Sachin Bhagchandani
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
33
|
Li H, He J, Zhang M, Liu J, Ni P. Glucose-Sensitive Polyphosphoester Diblock Copolymer for an Insulin Delivery System. ACS Biomater Sci Eng 2020; 6:1553-1564. [DOI: 10.1021/acsbiomaterials.9b01817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Hongping Li
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, P. R. China
| | - Jinlin He
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, P. R. China
| | - Mingzu Zhang
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, P. R. China
| | - Jian Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou 215123, P. R. China
| | - Peihong Ni
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
34
|
Muntoni E, Marini E, Ahmadi N, Milla P, Ghè C, Bargoni A, Capucchio MT, Biasibetti E, Battaglia L. Lipid nanoparticles as vehicles for oral delivery of insulin and insulin analogs: preliminary ex vivo and in vivo studies. Acta Diabetol 2019; 56:1283-1292. [PMID: 31407113 DOI: 10.1007/s00592-019-01403-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/06/2019] [Indexed: 12/18/2022]
Abstract
AIMS Subcutaneous administration of insulin in patients suffering from diabetes is associated with the distress of daily injections. Among alternative administration routes, the oral route seems to be the most advantageous for long-term administration, also because the peptide undergoes a hepatic first-pass effect, contributing to the inhibition of the hepatic glucose output. Unfortunately, insulin oral administration has so far been hampered by degradation by gastrointestinal enzymes and poor intestinal absorption. Loading in lipid nanoparticles should allow to overcome these limitations. METHODS Entrapment of peptides into such nanoparticles is not easy, because of their high molecular weight, hydrophilicity and thermo-sensitivity. In this study, this objective was achieved by employing fatty acid coacervation method: solid lipid nanoparticles and newly engineered nanostructured lipid carriers were formulated. Insulin and insulin analog-glargine insulin-were entrapped in the lipid matrix through hydrophobic ion pairing. RESULTS Bioactivity of lipid entrapped peptides was demonstrated through a suitable in vivo experiment. Ex vivo and in vivo studies were carried out by employing fluorescently labelled peptides. Gut tied up experiments showed the superiority of glargine insulin-loaded nanostructured lipid carriers, which demonstrated significantly higher permeation (till 30% dose/mL) compared to free peptide. Approximately 6% absolute bioavailability in the bloodstream was estimated for the same formulation through in vivo pharmacokinetic studies in rats. Consequently, a discrete blood glucose responsivity was noted in healthy animals. CONCLUSIONS Given the optimized ex vivo and in vivo intestinal uptake of glargine insulin from nanostructured lipid carriers, further studies will be carried out on healthy and diabetic rat models in order to establish a glargine insulin dose-glucose response relation.
Collapse
Affiliation(s)
- Elisabetta Muntoni
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, Turin, Italy
| | - Elisabetta Marini
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, Turin, Italy
| | - Nahid Ahmadi
- Department of Chemistry, University of Sistan and Baluchistan, University Boulevard, Zahedan, Iran
| | - Paola Milla
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, Turin, Italy
| | - Corrado Ghè
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, Turin, Italy
| | - Alessandro Bargoni
- Dipartimento di Scienze della Sanità Pubblica e Pediatriche, Università degli Studi di Torino, Piazza Polonia 94, Turin, Italy
| | - Maria Teresa Capucchio
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, Largo Paolo Braccini 2, Grugliasco, Italy
| | - Elena Biasibetti
- Histopathology Department CIBA, Istituto Zooprofilattico Sperimentale del Piemonte, Via Bologna 148, Turin, Italy
| | - Luigi Battaglia
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, Turin, Italy.
| |
Collapse
|
35
|
Wang J, Wang Z, Yu J, Zhang Y, Zeng Y, Gu Z. A forskolin-conjugated insulin analog targeting endogenous glucose-transporter for glucose-responsive insulin delivery. Biomater Sci 2019; 7:4508-4513. [PMID: 31608343 PMCID: PMC7148115 DOI: 10.1039/c9bm01283d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Insulin administration for the management of diabetes is accompanied by hypoglycemia, which is expected to be mitigated by glucose-responsive smart insulin that has self-regulation ability in response to blood glucose level (BGL) fluctuation. Here, we have prepared a new insulin analog by modifying insulin with forskolin (designated as insulin-F), a glucose-transporter (Glut) inhibitor. In vitro, insulin-F is capable of binding to Glut on erythrocyte ghosts, which can be inhibited by glucose and cytochalasin B. Upon subcutaneous injection in type 1 diabetic mice, insulin-F maintains BGLs below 200 mg mL-1 for up to 10 h, and achieves 20 h with two sequential injections. Moreover, insulin-F also binds to endogenous Gluts. Upon a glucose challenge, the elevated level of glucose competitively replaces and liberates insulin-F that binds to Glut, rapidly restoring BGLs to the normal range.
Collapse
Affiliation(s)
- Jinqiang Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA. and California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA. and California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Jicheng Yu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27514, USA
| | - Yuqi Zhang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27514, USA
| | - Yi Zeng
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA. and California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA. and California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90024, USA and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
36
|
|