1
|
Alizadehmojarad AA, Yang S, Gong X, Strano MS. Analysis of Glucose Responsive Glucagon Therapeutics using Computational Models of the Glucoregulatory System. Adv Healthc Mater 2024; 13:e2401410. [PMID: 39205540 PMCID: PMC11582512 DOI: 10.1002/adhm.202401410] [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: 04/17/2024] [Revised: 07/22/2024] [Indexed: 09/04/2024]
Abstract
Glucose-responsive glucagon (GRG) therapeutics are a promising technology for reducing the risk of severe hypoglycemia as a complication of diabetes mellitus. Herein, the performance of candidate GRGs in the literature by modeling the kinetics of activation and connecting them as input into physiological glucoregulatory models is evaluated and projected the two distinct GRG designs, experimental results reported in Wu et al. (GRG-I) and Webber et al. (GRG-II) is considered. Both are evaluated using a multi-compartmental glucoregulatory model (IMPACT) and used to compare in-vivo experimental data of therapeutic performance in rats and mice. For GRG-I and GRG-II, the total integrated glucose material balances are overestimated by 41.5% ± 14% and underestimated by 24.8% ± 16% compared to in-vivo time-course data, respectively. These large differences to the relatively simple computational descriptions of glucagon dynamics in the model, which underscores the urgent need for improved glucagon models is attributed. Additionally, therapeutic insulin and glucagon infusion pumps are modeled for type 1 diabetes mellitus (T1DM) human subjects to extend the results to additional datasets. These observations suggest that both the representative physiological and non-physiological models considered in this work require additional refinement to successfully describe clinical data that involve simultaneous, coupled insulin, glucose, and glucagon dynamics.
Collapse
Affiliation(s)
- Ali A Alizadehmojarad
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sungyun 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 S Strano
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
2
|
Allamreddy S, Arora M, Ganugula R, Friend R, Basu R, Kumar MNVR. Prospects for the convergence of polyphenols with pharmaceutical drugs in Type 2 Diabetes: challenges, risks, and strategies. Pharmacol Rev 2024; 77:PHARMREV-AR-2023-001074. [PMID: 39326899 DOI: 10.1124/pharmrev.124.001074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex disease that can lead to a variety of life-threatening secondary health conditions. Current treatment strategies primarily revolve around tight glucose control that is difficult to achieve and often turns out to be dangerous due to possible hypoglycemic events. Numerous long-term studies have demonstrated that complex pathways, including low-grade inflammation due to fluctuating glucose levels, are involved in the progression of the disease and the development of secondary health conditions. Growing clinical evidence supports the effectiveness of using multiple medications, possibly in combination with insulin, to effectively manage T2DM. On the other hand, despite the huge, largely untapped potential therapeutic benefit of 'polyphenols', there remains a general skepticism of the practice. However, for any evidence-based clinical intervention, the balance of benefits and risks takes center stage and is governed by biopharmaceutics principles. In this article, we outline the current clinical perspectives on pharmaceutical drug combinations, rationale for early initiation of insulin, and the advantages of novel dosage forms to meet the pathophysiological changes of T2DM, emphasizing the need for further clinical studies to substantiate these approaches. We also make the case for traditional medicines and their combinations with pharmaceutical drugs and outline the inherent challenges in doing so, while also providing recommendations for future research and clinical practice. Significance Statement Type 2 diabetes is associated with life-threatening secondary health conditions that are often difficult to treat. This review provides an in-depth account of preventing/delaying secondary health conditions through combination therapies and emphasizes the role of effective delivery strategies in realizing the translation of such combinations. We will build the case for the importance of polyphenols in diabetes, determine the reasons for skepticism, and potential combinations with pharmaceutical drugs.
Collapse
Affiliation(s)
| | - M Arora
- The University of Alabama, United States
| | - R Ganugula
- CCHS, The University of Alabama, United States
| | - R Friend
- The University of Alabama, United States
| | - R Basu
- Division of Endocrinology, Diabetes, and Metabolism, The University of Alabama at Birmingham, United States
| | - M N V Ravi Kumar
- Bioscience and Medicine, The University of Alabama, United States
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Xian S, Xiang Y, Liu D, Fan B, Mitrová K, Ollier RC, Su B, Alloosh MA, Jiráček J, Sturek M, Alloosh M, Webber MJ. Insulin-Dendrimer Nanocomplex for Multi-Day Glucose-Responsive Therapy in Mice and Swine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308965. [PMID: 37994248 DOI: 10.1002/adma.202308965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/27/2023] [Indexed: 11/24/2023]
Abstract
The management of diabetes in a manner offering autonomous insulin therapy responsive to glucose-directed need, and moreover with a dosing schedule amenable to facile administration, remains an ongoing goal to improve the standard of care. While basal insulins with reduced dosing frequency, even once-weekly administration, are on the horizon, there is still no approved therapy that offers glucose-responsive insulin function. Herein, a nanoscale complex combining both electrostatic- and dynamic-covalent interactions between a synthetic dendrimer carrier and an insulin analogue modified with a high-affinity glucose-binding motif yields an injectable insulin depot affording both glucose-directed and long-lasting insulin availability. Following a single injection, it is even possible to control blood glucose for at least one week in diabetic swine subjected to daily oral glucose challenges. Measurements of serum insulin concentration in response to challenge show increases in insulin corresponding to elevated blood glucose levels, an uncommon finding even in preclinical work on glucose-responsive insulin. Accordingly, the subcutaneous nanocomplex that results from combining electrostatic- and dynamic-covalent interactions between a modified insulin and a synthetic dendrimer carrier affords a glucose-responsive insulin depot for week-long control following a single routine injection.
Collapse
Affiliation(s)
- Sijie Xian
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Yuanhui Xiang
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Dongping Liu
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Bowen Fan
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Katarína Mitrová
- Czech Academy of Sciences, Institute of Organic Chemistry and Biochemistry, Prague, 16610, Czech Republic
| | - Rachel C Ollier
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Bo Su
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| | | | - Jiří Jiráček
- Czech Academy of Sciences, Institute of Organic Chemistry and Biochemistry, Prague, 16610, Czech Republic
| | | | | | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, 105 McCourtney Hall, Notre Dame, IN, 46556, USA
| |
Collapse
|
6
|
Zhang Y, Hung-Chieh Chou D. From Natural Insulin to Designed Analogs: A Chemical Biology Exploration. Chembiochem 2023; 24:e202300470. [PMID: 37800626 DOI: 10.1002/cbic.202300470] [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: 06/23/2023] [Revised: 08/25/2023] [Indexed: 10/07/2023]
Abstract
Since its discovery in 1921, insulin has been at the forefront of scientific breakthroughs. From its amino acid sequencing to the revelation of its three-dimensional structure, the progress in insulin research has spurred significant therapeutic breakthroughs. In recent years, protein engineering has introduced innovative chemical and enzymatic methods for insulin modification, fostering the development of therapeutics with tailored pharmacological profiles. Alongside these advances, the quest for self-regulated, glucose-responsive insulin remains a holy grail in the field. In this article, we highlight the pivotal role of chemical biology in driving these innovations and discuss how it continues to shape the future trajectory of insulin research.
Collapse
Affiliation(s)
- Yanxian Zhang
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, 1701 Page Mill Road, Palo Alto, CA 94304, USA
| | - Danny Hung-Chieh Chou
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, 1701 Page Mill Road, Palo Alto, CA 94304, USA
| |
Collapse
|
7
|
Zhang Y, Wu BM. Current Advances in Stimuli-Responsive Hydrogels as Smart Drug Delivery Carriers. Gels 2023; 9:838. [PMID: 37888411 PMCID: PMC10606589 DOI: 10.3390/gels9100838] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 10/28/2023] Open
Abstract
In recent years, significant advancements in the field of advanced materials and hydrogel engineering have enabled the design and fabrication of smart hydrogels and nanogels that exhibit sensitivity to specific signals or pathological conditions, leading to a wide range of applications in drug delivery and disease treatment. This comprehensive review aims to provide an in-depth analysis of the stimuli-responsive principles exhibited by smart hydrogels in response to various triggers, such as pH levels, temperature fluctuations, light exposure, redox conditions, or the presence of specific biomolecules. The functionality and performance characteristics of these hydrogels are highly influenced by both their constituent components and fabrication processes. Key design principles, their applications in disease treatments, challenges, and future prospects were also discussed. Overall, this review aims to contribute to the current understanding of gel-based drug delivery systems and stimulate further research in this rapidly evolving field.
Collapse
Affiliation(s)
- Yulong Zhang
- Department of Mineralized Tissue Biology, The Forsyth Institute, Cambridge, MA 02140, USA;
| | - Benjamin M. Wu
- Department of Mineralized Tissue Biology, The Forsyth Institute, Cambridge, MA 02140, USA;
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Cambuli VM, Baroni MG. Intelligent Insulin vs. Artificial Intelligence for Type 1 Diabetes: Will the Real Winner Please Stand Up? Int J Mol Sci 2023; 24:13139. [PMID: 37685946 PMCID: PMC10488097 DOI: 10.3390/ijms241713139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Research in the treatment of type 1 diabetes has been addressed into two main areas: the development of "intelligent insulins" capable of auto-regulating their own levels according to glucose concentrations, or the exploitation of artificial intelligence (AI) and its learning capacity, to provide decision support systems to improve automated insulin therapy. This review aims to provide a synthetic overview of the current state of these two research areas, providing an outline of the latest development in the search for "intelligent insulins," and the results of new and promising advances in the use of artificial intelligence to regulate automated insulin infusion and glucose control. The future of insulin treatment in type 1 diabetes appears promising with AI, with research nearly reaching the possibility of finally having a "closed-loop" artificial pancreas.
Collapse
Affiliation(s)
- Valentina Maria Cambuli
- Diabetology and Metabolic Diseaseas, San Michele Hospital, ARNAS Giuseppe Brotzu, 09121 Cagliari, Italy;
| | - Marco Giorgio Baroni
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Neuroendocrinology and Metabolic Diseases, IRCCS Neuromed, 86077 Pozzilli, Italy
| |
Collapse
|
11
|
Domingo-Lopez DA, Lattanzi G, H. J. Schreiber L, Wallace EJ, Wylie R, O'Sullivan J, Dolan EB, Duffy GP. Medical devices, smart drug delivery, wearables and technology for the treatment of Diabetes Mellitus. Adv Drug Deliv Rev 2022; 185:114280. [PMID: 35405298 DOI: 10.1016/j.addr.2022.114280] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/21/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus refers to a group of metabolic disorders which affect how the body uses glucose impacting approximately 9% of the population worldwide. This review covers the most recent technological advances envisioned to control and/or reverse Type 1 diabetes mellitus (T1DM), many of which will also prove effective in treating the other forms of diabetes mellitus. Current standard therapy for T1DM involves multiple daily glucose measurements and insulin injections. Advances in glucose monitors, hormone delivery systems, and control algorithms generate more autonomous and personalised treatments through hybrid and fully automated closed-loop systems, which significantly reduce hypo- and hyperglycaemic episodes and their subsequent complications. Bi-hormonal systems that co-deliver glucagon or amylin with insulin aim to reduce hypoglycaemic events or increase time spent in target glycaemic range, respectively. Stimuli responsive materials for the controlled delivery of insulin or glucagon are a promising alternative to glucose monitors and insulin pumps. By their self-regulated mechanism, these "smart" drugs modulate their potency, pharmacokinetics and dosing depending on patients' glucose levels. Islet transplantation is a potential cure for T1DM as it restores endogenous insulin and glucagon production, but its use is not yet widespread due to limited islet sources and risks of chronic immunosuppression. New encapsulation strategies that promote angiogenesis and oxygen delivery while protecting islets from recipients' immune response may overcome current limiting factors.
Collapse
|
12
|
Jarosinski MA, Chen YS, Varas N, Dhayalan B, Chatterjee D, Weiss MA. New Horizons: Next-Generation Insulin Analogues: Structural Principles and Clinical Goals. J Clin Endocrinol Metab 2022; 107:909-928. [PMID: 34850005 PMCID: PMC8947325 DOI: 10.1210/clinem/dgab849] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Indexed: 11/19/2022]
Abstract
Design of "first-generation" insulin analogues over the past 3 decades has provided pharmaceutical formulations with tailored pharmacokinetic (PK) and pharmacodynamic (PD) properties. Application of a molecular tool kit-integrating protein sequence, chemical modification, and formulation-has thus led to improved prandial and basal formulations for the treatment of diabetes mellitus. Although PK/PD changes were modest in relation to prior formulations of human and animal insulins, significant clinical advantages in efficacy (mean glycemia) and safety (rates of hypoglycemia) were obtained. Continuing innovation is providing further improvements to achieve ultrarapid and ultrabasal analogue formulations in an effort to reduce glycemic variability and optimize time in range. Beyond such PK/PD metrics, next-generation insulin analogues seek to exploit therapeutic mechanisms: glucose-responsive ("smart") analogues, pathway-specific ("biased") analogues, and organ-targeted analogues. Smart insulin analogues and delivery systems promise to mitigate hypoglycemic risk, a critical barrier to glycemic control, whereas biased and organ-targeted insulin analogues may better recapitulate physiologic hormonal regulation. In each therapeutic class considerations of cost and stability will affect use and global distribution. This review highlights structural principles underlying next-generation design efforts, their respective biological rationale, and potential clinical applications.
Collapse
Affiliation(s)
- Mark A Jarosinski
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nicolás Varas
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Deepak Chatterjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
13
|
Pissarnitski DA, Kekec A, Yan L, Zhu Y, Feng DD, Huo P, Madsen-Duggan C, Moyes CR, Nargund RP, Kelly T, Zhang X, Carballo-Jane E, Gorski J, Zafian P, Qatanani M, Kaarsholm N, Meng F, Jia X, Lee KJ, Wang W, Xu S, Hohn MJ, Iammarino MJ, McCoy MA, Okoh GA, Liang Y, Hollingsworth SA, Erion MD, Kelley DE, Garbaccio RM, Zhang A, Mu J, Lin S. Discovery of Insulin Receptor Partial Agonists MK-5160 and MK-1092 as Novel Basal Insulins with Potential to Improve Therapeutic Index. J Med Chem 2022; 65:5593-5605. [PMID: 35298158 DOI: 10.1021/acs.jmedchem.1c02073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We have identified a series of novel insulin receptor partial agonists (IRPAs) with a potential to mitigate the risk of hypoglycemia associated with the use of insulin as an antidiabetic treatment. These molecules were designed as dimers of native insulin connected via chemical linkers of variable lengths with optional capping groups at the N-terminals of insulin chains. Depending on the structure, the maximal activation level (%Max) varied in the range of ∼20-70% of native insulin, and EC50 values remained in sub-nM range. Studies in minipig and dog demonstrated that IRPAs had sufficient efficacy to normalize plasma glucose levels in diabetes, while providing reduction of hypoglycemia risk. IRPAs had a prolonged duration of action, potentially making them suitable for once-daily dosing. Two lead compounds with %Max values of 30 and 40% relative to native insulin were selected for follow up studies in the clinic.
Collapse
Affiliation(s)
| | - Ahmet Kekec
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Lin Yan
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Yuping Zhu
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Danqing D Feng
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Pei Huo
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | | | - Ravi P Nargund
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Terri Kelly
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Xiaoping Zhang
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | - Judith Gorski
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Peter Zafian
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mo Qatanani
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Niels Kaarsholm
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Fanyu Meng
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Xiujuan Jia
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Keun-Joong Lee
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Weixun Wang
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Sherrie Xu
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Michael J Hohn
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | - Mark A McCoy
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Grace A Okoh
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yingkai Liang
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Mark D Erion
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - David E Kelley
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | - Amy Zhang
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - James Mu
- Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Songnian Lin
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| |
Collapse
|
14
|
Abstract
Insulin therapy has a long history at the cutting edge of technological development through purification, extended-action, molecular chemistry, and devices, and in support technologies including self-measurement and patient education. But unmet needs remain large. Today's therapy cannot deliver minute-to-minute control of glucose levels, and cannot imitate the reflex/incretin driven physiological insulin delivery at mealtimes. Further it depends on a raft of devices for administration several times a day, devices liked for their functionality, but disliked as an intrusive reminder of the condition, several times a day. Approaches to overcoming these barriers include closed-loop systems and further modification of insulin formulations, but are limited by fundamental underlying difficulties. While clinical studies of oral insulin are in progress, the barriers to success look daunting. Development of small-molecule approaches (insulin-mimetic tablets) appears to have stalled, while concepts for glucose-responsive insulin as yet fail to deliver the necessary insulin-to-glucose gradient. Gene therapy, feasible in animals in preliminary studies, is not capable of providing feedback control. Transplantation of cultured islets and islet B-cells from stem cells thus looks to the be the best long-term prospect for insulin delivery in terms of overcoming the above barriers, but is a true biotechnological tour-de-force which will take time to mature.
Collapse
Affiliation(s)
- Philip Home
- Translational and Clinical Research Institute, Newcastle University, UK.
| |
Collapse
|
15
|
Home PD, Mehta R. Insulin therapy development beyond 100 years. Lancet Diabetes Endocrinol 2021; 9:695-707. [PMID: 34480874 DOI: 10.1016/s2213-8587(21)00182-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022]
Abstract
The first insulin preparation capable of consistently lowering blood glucose was developed in 1921. But 100 years later, blood glucose control with insulin in people with diabetes is nearly universally suboptimal, with essentially the same molecule still delivered by the same inappropriate subcutaneous injection route. Bypassing this route with oral administration appears to have become technologically feasible, accelerating over the past 50 years, either with packaged insulin peptides or by chemical insulin mimetics. Some of the problems of prospective unregulated absorption of insulin into the circulation from subcutaneous depots might be overcome with glucose-responsive insulins. Approaches to these problems could be modification of the peptide by adducts, or the use of nanoparticles or insulin patches, which deliver insulin according to glucose concentration. Some attention has been paid to targeting insulin preferentially to different organs, either by molecular engineering of insulin, or with adducts. But all these approaches still have problems in even beginning to match the responsiveness of physiological insulin delivery to metabolic requirements, both prandially and basally. As would be expected, for all these technically complex approaches, many examples of abandoned development can be found. Meanwhile, it is becoming possible to change the duration of action of subcutaneous injected insulin analogues to act even more rapidly for meals, and towards weekly insulin for basal administration. The state of the art of all these approaches, and the barriers to success, are reviewed here.
Collapse
Affiliation(s)
- Philip D Home
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| | - Roopa Mehta
- Metabolic Diseases Research Unit, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
| |
Collapse
|
16
|
Nauck MA, Wefers J, Meier JJ. Treatment of type 2 diabetes: challenges, hopes, and anticipated successes. Lancet Diabetes Endocrinol 2021; 9:525-544. [PMID: 34181914 DOI: 10.1016/s2213-8587(21)00113-3] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
Despite the successful development of new therapies for the treatment of type 2 diabetes, such as glucagon-like peptide-1 (GLP-1) receptor agonists and sodium-glucose cotransporter-2 inhibitors, the search for novel treatment options that can provide better glycaemic control and at reduce complications is a continuous effort. The present Review aims to present an overview of novel targets and mechanisms and focuses on glucose-lowering effects guiding this search and developments. We discuss not only novel developments of insulin therapy (eg, so-called smart insulin preparation with a glucose-dependent mode of action), but also a group of drug classes for which extensive research efforts have not been rewarded with obvious clinical impact. We discuss the potential clinical use of the salutary adipokine adiponectin and the hepatokine fibroblast growth factor (FGF) 21, among others. A GLP-1 peptide receptor agonist (semaglutide) is now available for oral absorption, and small molecules activating GLP-1 receptors appear on the horizon. Bariatric surgery and its accompanying changes in the gut hormonal milieu offer a background for unimolecular peptides interacting with two or more receptors (for GLP-1, glucose-dependent insulinotropic polypeptide, glucagon, and peptide YY) and provide more substantial glycaemic control and bodyweight reduction compared with selective GLP-1 receptor agonists. These and additional approaches will help expand the toolbox of effective medications needed for optimising the treatment of well delineated subgroups of type 2 diabetes or help develop personalised approaches for glucose-lowering drugs based on individual characteristics of our patients.
Collapse
Affiliation(s)
- Michael A Nauck
- Diabetes Division, Katholisches Klinikum Bochum, St Josef Hospital, Ruhr University Bochum, Bochum, Germany.
| | - Jakob Wefers
- Diabetes Division, Katholisches Klinikum Bochum, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Juris J Meier
- Diabetes Division, Katholisches Klinikum Bochum, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| |
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: 5.0] [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
|
Abstract
Although insulin therapy was already introduced one-hundred years ago, insulin formulations are still being refined to reduce the risk of hypoglycaemia and of other insulin side effects such as weight gain. This review summarises the available clinical data for some ongoing developments of new insulins and evaluates their potential for future insulin therapy. Once-weekly insulins will most likely be the next addition to the insulin armamentarium. First clinical studies indicate low peak-to-trough fluctuations with these insulins indicating the potential to achieve better glycaemic control or reduce hypoglycaemic events versus available basal insulins. Proof-of-concept has also been established for hepato-preferential and oral insulins; however, adverse effects and low bioavailability still need to be overcome. It will take much longer, before glucose-responsive "smart" insulins will be available. A first clinical study and numerous pre-clinical data show the potential, but also the challenges of designing an insulin that quickly reacts to blood glucose changes and prevents hypoglycaemia and pronounced hyperglycaemia. Nevertheless, it is reassuring that the search for better insulins has never stopped since its first use one-hundred years ago and is still ongoing. New developments have a high potential of further improving the safety and efficacy of insulin therapy in the future.
Collapse
|
19
|
Fuchs S, Ernst AU, Wang LH, Shariati K, Wang X, Liu Q, Ma M. Hydrogels in Emerging Technologies for Type 1 Diabetes. Chem Rev 2020; 121:11458-11526. [DOI: 10.1021/acs.chemrev.0c01062] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Stephanie Fuchs
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Alexander U. Ernst
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Long-Hai Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Kaavian Shariati
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Xi Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Qingsheng Liu
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Minglin Ma
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
20
|
Abstract
Insulin therapy has advanced remarkably over the past few decades. Ultra-rapid-acting and ultra-long-acting insulin analogs are now commercially available. Many additional insulin formulations are in development. This review outlines recent advances in insulin therapy and novel therapies in development.
Collapse
Affiliation(s)
- Leah M. Wilson
- Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Jessica R. Castle
- Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, Portland, Oregon, USA
- Address correspondence to: Jessica R. Castle, MD, Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, L607, Portland, OR 97239-3098, USA
| |
Collapse
|
21
|
Affiliation(s)
- Simeon I Taylor
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | | |
Collapse
|
22
|
Yang JF, Gong X, Bakh NA, Carr K, Phillips NFB, Ismail-Beigi F, Weiss MA, Strano MS. Connecting Rodent and Human Pharmacokinetic Models for the Design and Translation of Glucose-Responsive Insulin. Diabetes 2020; 69:1815-1826. [PMID: 32152206 PMCID: PMC8176262 DOI: 10.2337/db19-0879] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/08/2020] [Indexed: 12/16/2022]
Abstract
Despite considerable progress, development of glucose-responsive insulins (GRIs) still largely depends on empirical knowledge and tedious experimentation-especially on rodents. To assist the rational design and clinical translation of the therapeutic, we present a Pharmacokinetic Algorithm Mapping GRI Efficacies in Rodents and Humans (PAMERAH) built upon our previous human model. PAMERAH constitutes a framework for predicting the therapeutic efficacy of a GRI candidate from its user-specified mechanism of action, kinetics, and dosage, which we show is accurate when checked against data from experiments and literature. Results from simulated glucose clamps also agree quantitatively with recent GRI publications. We demonstrate that the model can be used to explore the vast number of permutations constituting the GRI parameter space and thereby identify the optimal design ranges that yield desired performance. A design guide aside, PAMERAH more importantly can facilitate GRI's clinical translation by connecting each candidate's efficacies in rats, mice, and humans. The resultant mapping helps to find GRIs that appear promising in rodents but underperform in humans (i.e., false positives). Conversely, it also allows for the discovery of optimal human GRI dynamics not captured by experiments on a rodent population (false negatives). We condense such information onto a "translatability grid" as a straightforward, visual guide for GRI development.
Collapse
Affiliation(s)
- Jing Fan Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Xun Gong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Naveed A Bakh
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Kelley Carr
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH
| | | | | | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Michael S Strano
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
23
|
Abstract
BACKGROUND Insulin therapy is the mainstay of treatment for type 1 diabetes and may be necessary in type 2 diabetes. Current insulin analogues present a more physiological profile, are effective, and with less risk of hypoglycemia, but they are expensive. Biosimilar insulins should offer the advantages of insulin analogues at reduced costs. In addition, current rapid-acting insulin analogues are not fast enough to control excessive postprandial glucose excursions in many patients. AREAS OF UNCERTAINTY Biosimilar insulins demonstrated that are safe and effective, but interchangeability and automatic substitution remain an issue. Ultrafast-acting insulins should reduce postprandial hyperglycemia and improve flexibility in insulin dosing. DATA SOURCES This systematic review was conducted following widely recommended methods. We searched for each topic in Medline, Embase, the Cochrane Library, and SCISEARCH for relevant citations for the appropriate period. THERAPEUTIC ADVANCES LY2963016 and MK-1293 are biosimilar insulins of insulin glargine, and SAR342434 is a biosimilar of insulin lispro. The abbreviated developed program demonstrated comparable efficacy and safety and supports their use for treatment of people with diabetes but no interchangeability. Faster-acting insulin aspart is a new formulation of insulin aspart with accelerated subcutaneous absorption. Faster aspart demonstrated noninferiority in reducing HbA1c as compared to insulin aspart with superiority in controlling postprandial hyperglycemia without increasing hypoglycemia, and flexible insulin dosing. CONCLUSIONS Biosimilar insulins have comparable PK-PD profiles and equivalent efficacy and safety to original insulins at a lower price, making them available for more people with diabetes. Faster aspart is the first ultrafast-acting insulin. New upcoming clinical trials and more clinical experience with faster aspart will show the real potential of this new insulin.
Collapse
|
24
|
Primavera R, Kevadiya BD, Swaminathan G, Wilson RJ, De Pascale A, Decuzzi P, Thakor AS. Emerging Nano- and Micro-Technologies Used in the Treatment of Type-1 Diabetes. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E789. [PMID: 32325974 PMCID: PMC7221526 DOI: 10.3390/nano10040789] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
Type-1 diabetes is characterized by high blood glucose levels due to a failure of insulin secretion from beta cells within pancreatic islets. Current treatment strategies consist of multiple, daily injections of insulin or transplantation of either the whole pancreas or isolated pancreatic islets. While there are different forms of insulin with tunable pharmacokinetics (fast, intermediate, and long-acting), improper dosing continues to be a major limitation often leading to complications resulting from hyper- or hypo-glycemia. Glucose-responsive insulin delivery systems, consisting of a glucose sensor connected to an insulin infusion pump, have improved dosing but they still suffer from inaccurate feedback, biofouling and poor patient compliance. Islet transplantation is a promising strategy but requires multiple donors per patient and post-transplantation islet survival is impaired by inflammation and suboptimal revascularization. This review discusses how nano- and micro-technologies, as well as tissue engineering approaches, can overcome many of these challenges and help contribute to an artificial pancreas-like system.
Collapse
Affiliation(s)
- Rosita Primavera
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; (R.P.); (B.D.K.); (G.S.); (R.J.W.)
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Bhavesh D Kevadiya
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; (R.P.); (B.D.K.); (G.S.); (R.J.W.)
| | - Ganesh Swaminathan
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; (R.P.); (B.D.K.); (G.S.); (R.J.W.)
| | - Rudilyn Joyce Wilson
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; (R.P.); (B.D.K.); (G.S.); (R.J.W.)
| | - Angelo De Pascale
- Unit of Endocrinology, Department of Internal Medicine & Medical Specialist (DIMI), University of Genoa, 16163 Genoa, Italy;
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; (R.P.); (B.D.K.); (G.S.); (R.J.W.)
| |
Collapse
|
25
|
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: 123] [Impact Index Per Article: 24.6] [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
|
26
|
Abstract
Although insulin analogs have markedly improved glycemic control for people with diabetes, glycemic excursions still cause major health problems and complications. In particular, the narrow therapeutic window of current insulin therapy makes it extremely difficult to maintain normoglycemia without risking severe hypoglycemia. Currently, there are no FDA-approved insulin therapeutics whose bioactivity is regulated by blood glucose levels. This review discusses recent progress on developing glucose-responsive insulin (GRI) bioconjugates without the need of exogenous matrices. Through this approach, tremendous efforts have been made over the years to demonstrate the promise of better glycemic control and reduced risk of hypoglycemia. Last, we discuss future directions of GRI development with a goal to maximize the glucose responsiveness.
Collapse
Affiliation(s)
- Maria M. Disotuar
- Department of Biochemistry, University
of Utah, Salt Lake City, UT, USA
| | - Diao Chen
- Department of Biochemistry, University
of Utah, Salt Lake City, UT, USA
| | - Nai-Pin Lin
- Department of Biochemistry, University
of Utah, Salt Lake City, UT, USA
| | - Danny Hung-Chieh Chou
- Department of Biochemistry, University
of Utah, Salt Lake City, UT, USA
- Danny Hung-Chieh Chou, PhD, Department of
Biochemistry, University of Utah, 15 N Medical Drive East 4100, Salt Lake City
UT 84112, USA.
| |
Collapse
|
27
|
Visser SAG, Kandala B, Fancourt C, Krug AW, Cho CR. A Model-Informed Drug Discovery and Development Strategy for the Novel Glucose-Responsive Insulin MK-2640 Enabled Rapid Decision Making. Clin Pharmacol Ther 2020; 107:1296-1311. [PMID: 31889297 PMCID: PMC7325312 DOI: 10.1002/cpt.1729] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022]
Abstract
A model‐informed drug discovery and development strategy played a key role in the novel glucose‐responsive insulin MK‐2640’s early clinical development strategy and supported a novel clinical trial paradigm to assess glucose responsiveness. The development and application of in silico modeling approaches by leveraging substantial published clinical insulin pharmacokinetic–pharmacodynamic (PKPD) data and emerging preclinical and clinical data enabled rapid quantitative decision making. Learnings can be applied to define PKPD properties of novel insulins that could become therapeutically meaningful for diabetic patients.
Collapse
Affiliation(s)
- Sandra A G Visser
- Department of Quantitative Pharmacology & Pharmacometrics (QP2) at Merck & Co. Inc., Kenilworth, New Jersey, USA
| | - Bhargava Kandala
- Department of Quantitative Pharmacology & Pharmacometrics (QP2) at Merck & Co. Inc., Kenilworth, New Jersey, USA
| | - Craig Fancourt
- Department of Quantitative Pharmacology & Pharmacometrics (QP2) at Merck & Co. Inc., Kenilworth, New Jersey, USA
| | - Alexander W Krug
- Department of Translational Pharmacology at Merck & Co. Inc., Kenilworth, New Jersey, USA
| | - Carolyn R Cho
- Department of Quantitative Pharmacology & Pharmacometrics (QP2) at Merck & Co. Inc., Kenilworth, New Jersey, USA
| |
Collapse
|
28
|
Warshauer JT, Bluestone JA, Anderson MS. New Frontiers in the Treatment of Type 1 Diabetes. Cell Metab 2020; 31:46-61. [PMID: 31839487 PMCID: PMC6986815 DOI: 10.1016/j.cmet.2019.11.017] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/08/2019] [Accepted: 11/18/2019] [Indexed: 12/30/2022]
Abstract
Type 1 diabetes is an autoimmune disease caused by the immune-mediated destruction of pancreatic β cells that results in lifelong absolute insulin deficiency. For nearly a century, insulin replacement has been the only therapy for most people living with this disease. Recent advances in technology and our understanding of β cell development, glucose metabolism, and the underlying immune pathogenesis of the disease have led to innovative therapeutic and preventative approaches. A paradigm shift in immunotherapy development toward the targeting of islet-specific immune pathways involved in tolerance has driven the development of therapies that may allow for the prevention or reversal of this disease while avoiding toxicities associated with historical approaches that were broadly immunosuppressive. In this review, we discuss successes, failures, and emerging pharmacological therapies for type 1 diabetes that are changing how we approach this disease, from improving glycemic control to developing the "holy grail" of disease prevention.
Collapse
Affiliation(s)
- Jeremy T Warshauer
- Endocrine Division, Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey A Bluestone
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Mark S Anderson
- Endocrine Division, Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
29
|
|