1
|
Li D, Qin Q, Benetti AA, Kahouadji L, Wacker MG. BioJect: An in vitro platform to explore release dynamics of peptides in subcutaneous drug delivery. J Control Release 2025; 380:1058-1079. [PMID: 39923852 DOI: 10.1016/j.jconrel.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Predicting the release performance of subcutaneous (SC) drug formulations is challenging due to the complex interplay between physicochemical properties and the physiological microenvironment, which includes the extracellular matrix (ECM), fluid composition, and fluid availability, factors that collectively influence bioavailability and absorption rates. The ECM often acts as a bandpass filter modulated by local ion and protein content. In this study, we introduce the BioJect cell, a modern release test method based on the compendial flow-through cell, integrating a perfusion system with customizable biomatrix components. We systematically investigated the release mechanisms of four insulin formulations: regular human insulin, insulin aspart, insulin glulisine, and Neutral Protamine Hagedorn (NPH) insulin. A modified simulated subcutaneous interstitial fluid (mSSIF) comprising multiple components of the SC physiological environment was employed. It incorporates important ions and proteins (138.5 mM sodium, 10 mM potassium, 1.8 mM calcium, 0.8 mM magnesium, 111.3 mM chloride, 28 mM bicarbonate, 0.5 mM sulfate, 5 mM acetate, 4.2 mM phosphate, 30 g/L total protein added as bovine serum albumin). Our release test method discriminated the tested formulations under varying biorelevant conditions, demonstrating its biopredictive capabilities. Notably, we discovered a previously undocumented albumin binding affecting the release rate of insulin glulisine, likely occurring in the low-shear environment of SC tissue only. Additionally, the inclusion of biorelevant components like hyaluronic acid and collagen into the biomatrix of the BioJect cell provided profound insights into potential absorption and release mechanisms, supported by two in vitro-in vivo relationships (level C and level A). The BioJect cell represents a significant advancement in simulating the SC environment for drug release testing. Our findings highlight the importance of considering protein binding and ECM components in predicting drug absorption, offering a promising tool for the development and optimization of SC formulations.
Collapse
Affiliation(s)
- David Li
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore
| | - Qiuhua Qin
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore
| | - Ayça Altay Benetti
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore
| | - Lyes Kahouadji
- Department of Chemical Engineering, Imperial College London, London, United Kingdom
| | - Matthias G Wacker
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore.
| |
Collapse
|
2
|
He L, Jiang B, Peng Y, Zhang X, Liu M. NMR Based Methods for Metabolites Analysis. Anal Chem 2025; 97:5393-5406. [PMID: 40048643 PMCID: PMC11923949 DOI: 10.1021/acs.analchem.4c06477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/07/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Metabolite analysis is essential for understanding the biochemical processes and pathways that sustain life, providing insights into the complex interactions within cellular systems and clinical examinations. This review explores recent applications of nuclear magnetic resonance (NMR) spectroscopy in metabolite studies. Various methods enhancing analytical accuracy for metabolome profiling and metabolic pathway studies, including spectral simplification techniques, quantitative NMR, high-resolution MAS NMR, and isotopic labeling, are discussed. The application of NMR in in situ and in vivo studies is also covered, highlighting in-cell NMR and in vivo MRS techniques. Last but not least, we discuss recent advancements in NMR hyperpolarization, with a focus on dynamic nuclear polarization (DNP), chemically induced dynamic nuclear polarization (CIDNP), para-hydrogen-induced polarization (PHIP), and signal amplification by reversible exchange (SABRE). These advancements offer significant potential for enhancing the sensitivity and accuracy of metabolite studies and are expected to further deepen the study and understanding of metabolites and metabolic pathways.
Collapse
Affiliation(s)
- Lichun He
- State
Key Laboratory of Magnetic Resonance and Atomic Molecular Physics,
National Center for Magnetic Resonance in Wuhan, Innovation Academy
for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University
of Chinese Academy of Sciences, Beijing 101408, China
| | - Bin Jiang
- State
Key Laboratory of Magnetic Resonance and Atomic Molecular Physics,
National Center for Magnetic Resonance in Wuhan, Innovation Academy
for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University
of Chinese Academy of Sciences, Beijing 101408, China
| | - Yun Peng
- State
Key Laboratory of Magnetic Resonance and Atomic Molecular Physics,
National Center for Magnetic Resonance in Wuhan, Innovation Academy
for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University
of Chinese Academy of Sciences, Beijing 101408, China
| | - Xu Zhang
- State
Key Laboratory of Magnetic Resonance and Atomic Molecular Physics,
National Center for Magnetic Resonance in Wuhan, Innovation Academy
for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University
of Chinese Academy of Sciences, Beijing 101408, China
| | - Maili Liu
- State
Key Laboratory of Magnetic Resonance and Atomic Molecular Physics,
National Center for Magnetic Resonance in Wuhan, Innovation Academy
for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University
of Chinese Academy of Sciences, Beijing 101408, China
- Optics
Valley Laboratory, Hubei 430074, China
| |
Collapse
|
3
|
Giorgino F, Battelino T, Bergenstal RM, Forst T, Green JB, Mathieu C, Rodbard HW, Schnell O, Wilmot EG. The Role of Ultra-Rapid-Acting Insulin Analogs in Diabetes: An Expert Consensus. J Diabetes Sci Technol 2025; 19:452-469. [PMID: 37937585 PMCID: PMC11874134 DOI: 10.1177/19322968231204584] [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] [Indexed: 11/09/2023]
Abstract
Ultra-rapid-acting insulin analogs (URAA) are a further development and refinement of rapid-acting insulin analogs. Because of their adapted formulation, URAA provide an even faster pharmacokinetics and thus an accelerated onset of insulin action than conventional rapid-acting insulin analogs, allowing for a more physiologic delivery of exogenously applied insulin. Clinical trials have confirmed the superiority of URAA in controlling postprandial glucose excursions, with a safety profile that is comparable to the rapid-acting insulins. Consequently, many individuals with diabetes mellitus may benefit from URAA in terms of prandial glycemic control. Unfortunately, there are only few available recommendations from authoritative sources for use of URAA in clinical practice. Therefore, this expert consensus report aims to define populations of people with diabetes mellitus for whom URAA may be beneficial and to provide health care professionals with concrete, practical recommendations on how best to use URAA in this context.
Collapse
Affiliation(s)
- Francesco Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | - Tadej Battelino
- Department of Endocrinology, Diabetes and Metabolism, UCH-University Medical Center Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Thomas Forst
- Department of Endocrinology and Metabolic Diseases, Johannes Gutenberg University Medical Center, Mainz, Germany
- Clinical Research Services, Mannheim, Germany
| | - Jennifer B. Green
- Division of Endocrinology and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, University Hospital Gasthuisberg, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | - Oliver Schnell
- Forschergruppe Diabetes eV at the Helmholtz Centre, Munich-Neuherberg, Germany
| | - Emma G. Wilmot
- Department of Diabetes & Endocrinology, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
- Academic Unit for Translational Medical Sciences, University of Nottingham, Nottingham, England, UK
| |
Collapse
|
4
|
Emad-Eldin M, Balata GF, Elshorbagy EA, Hamed MS, Attia MS. Insulin therapy in type 2 diabetes: Insights into clinical efficacy, patient-reported outcomes, and adherence challenges. World J Diabetes 2024; 15:828-852. [PMID: 38766443 PMCID: PMC11099362 DOI: 10.4239/wjd.v15.i5.828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/01/2024] [Accepted: 03/20/2024] [Indexed: 05/10/2024] Open
Abstract
Insulin therapy plays a crucial role in the management of type 2 diabetes as the disease progresses. Over the past century, insulin formulations have undergone significant modifications and bioengineering, resulting in a diverse range of available insulin products. These products show distinct pharmacokinetic and pharmacodynamic profiles. Consequently, various insulin regimens have em-erged for the management of type 2 diabetes, including premixed formulations and combinations of basal and bolus insulins. The utilization of different insulin regimens yields disparate clinical outcomes, adverse events, and, notably, patient-reported outcomes (PROs). PROs provide valuable insights from the patient's perspective, serving as a valuable mine of information for enhancing healthcare and informing clinical decisions. Adherence to insulin therapy, a critical patient-reported outcome, significantly affects clinical outcomes and is influenced by multiple factors. This review provides insights into the clinical effectiveness of various insulin preparations, PROs, and factors impacting insulin therapy adherence, with the aim of enhancing healthcare practices and informing clinical decisions for individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Mahmoud Emad-Eldin
- Department of Pharmacy Practice, Faculty of Pharmacy, Zagazig University, Zagazig HFQM+872, Al-Sharqia Governorate, Egypt
| | - Gehan F Balata
- Department of Pharmacy Practice, Faculty of Pharmacy, Heliopolis University, Cairo 44519, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Al-Sharqia Governorate, Egypt
| | - Eman A Elshorbagy
- Department of Internal Medicine, Faculty of Medicine, Zagazig University, Zagazig 44519, Al-Sharqia Governorate, Egypt
| | - Mona S Hamed
- Department of Community at Faculty of Medicine, Zagazig University, Zagazig 44519, Al-Sharqia Governorate, Egypt
| | - Mohamed S Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Al-Sharqia Governorate, Egypt
| |
Collapse
|
5
|
Schöner TA, Vogel V, Venczel M, Knoth K, Kamm W, Paehler T, Louit G, Terán IT, Mundinger P, Marker A, Loos P, Hittinger M, Lehr CM. Biorelevant subcutaneous in vitro test predicts the release of human and fast acting insulin formulations. Int J Pharm 2024; 655:123995. [PMID: 38490402 DOI: 10.1016/j.ijpharm.2024.123995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024]
Abstract
The administration of insulins by subcutaneous injection is nowadays widely prevalent. The injection site is located below the dermis and composed of cells and the extracellular matrix formed of a network of macromolecules such as hyaluronic acid and collagen. Following an injection, the insulins from the formulated products are timely released as drug molecules from the injection site into systemic circulation. In this publication, we show the development of an in vitro setup utilizing a hydrogel composed of a special collagen-hyaluronic acid mixture that mimics the extracellular matrix. Another setup was used for differentiation of the commercially available and research insulin formulations by determining the in vitro permeation characteristics with the results that were correlated with the human in vivo data. Significant differentiation was achieved at 90 % confidence level between the permeation curves of insulin glulisine containing formulations (U100 and a concentrated research formulation), while in case of the insulin lispro containing formulations (U100 and U200) the permeation curves showed similarity. These results demonstrated that the in vitro setup may be used as a tool for formulation development and drug candidate profiling as it is able to differentiate or show similarities between the agglomeration states and concentration of the active pharmaceutical ingredients.
Collapse
Affiliation(s)
| | | | - Marta Venczel
- Sanofi-Aventis Deutschland GmbH, Frankfurt/Main,Germany.
| | | | - Walter Kamm
- Sanofi-Aventis Deutschland GmbH, Frankfurt/Main,Germany
| | | | | | | | | | | | - Petra Loos
- Sanofi-Aventis Deutschland GmbH, Frankfurt/Main,Germany
| | | | - Claus-Michael Lehr
- PharmBioTec GmbH, 66123 Saarbruecken, Germany; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) and Saarland University, 66123 Saarbruecken, Germany
| |
Collapse
|
6
|
Uflewski M, Rindfleisch T, Korkmaz K, Tietz E, Mielke S, Correa Galvis V, Dünschede B, Luzarowski M, Skirycz A, Schwarzländer M, Strand DD, Hertle AP, Schünemann D, Walther D, Thalhammer A, Wolff M, Armbruster U. The thylakoid proton antiporter KEA3 regulates photosynthesis in response to the chloroplast energy status. Nat Commun 2024; 15:2792. [PMID: 38555362 PMCID: PMC10981695 DOI: 10.1038/s41467-024-47151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/19/2024] [Indexed: 04/02/2024] Open
Abstract
Plant photosynthesis contains two functional modules, the light-driven reactions in the thylakoid membrane and the carbon-fixing reactions in the chloroplast stroma. In nature, light availability for photosynthesis often undergoes massive and rapid fluctuations. Efficient and productive use of such variable light supply requires an instant crosstalk and rapid synchronization of both functional modules. Here, we show that this communication involves the stromal exposed C-terminus of the thylakoid K+-exchange antiporter KEA3, which regulates the ΔpH across the thylakoid membrane and therefore pH-dependent photoprotection. By combining in silico, in vitro, and in vivo approaches, we demonstrate that the KEA3 C-terminus senses the energy state of the chloroplast in a pH-dependent manner and regulates transport activity in response. Together our data pinpoint a regulatory feedback loop by which the stromal energy state orchestrates light capture and photoprotection via multi-level regulation of KEA3.
Collapse
Affiliation(s)
- Michał Uflewski
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Tobias Rindfleisch
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
- Department of Physical Biochemistry, University of Potsdam, D-14476, Potsdam, Germany
- Computational Biology Unit, Department of Chemistry, University of Bergen, Bergen, Norway
| | - Kübra Korkmaz
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Enrico Tietz
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Sarah Mielke
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Viviana Correa Galvis
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Beatrix Dünschede
- Molecular Biology of Plant Organelles, Faculty of Biology and Biotechnology, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Marcin Luzarowski
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Aleksandra Skirycz
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Markus Schwarzländer
- Institute of Plant Biology and Biotechnology (IBBP), Universität Münster, Schlossplatz 8, D-48143, Münster, Germany
| | - Deserah D Strand
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Alexander P Hertle
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Danja Schünemann
- Molecular Biology of Plant Organelles, Faculty of Biology and Biotechnology, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Dirk Walther
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany
| | - Anja Thalhammer
- Department of Physical Biochemistry, University of Potsdam, D-14476, Potsdam, Germany
| | - Martin Wolff
- Department of Physical Biochemistry, University of Potsdam, D-14476, Potsdam, Germany
| | - Ute Armbruster
- Max Planck Institute of Molecular Plant Physiology, Wissenschaftspark Golm, Potsdam, D-14476, Germany.
- Molecular Photosynthesis, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, D-40225, Düsseldorf, Germany.
- CEPLAS - Cluster of Excellence on Plant Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225, Düsseldorf, Germany.
| |
Collapse
|
7
|
Kommera SP, Kumar A, Chitkara D, Mittal A. Pramlintide an Adjunct to Insulin Therapy: Challenges and Recent Progress in Delivery. J Pharmacol Exp Ther 2024; 388:81-90. [PMID: 37863489 DOI: 10.1124/jpet.123.001679] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/22/2023] Open
Abstract
Dysregulation of various glucoregulatory hormones lead to failure of insulin monotherapy in patients with diabetes mellitus due to various reasons, including severe hypoglycemia, glycemic hypervariability, and an increased risk of microvascular complications. However, pramlintide as an adjunct to insulin therapy enhances glucagon suppression and thereby offers improved glycemic control. Clinical studies have shown that pramlintide improves glycemic control, reduces postprandial glucose excursions, and promotes weight loss in patients with type 1 and type 2 diabetes. Although clinical benefits of pramlintide are well reported, there still exists a high patient resistance for the therapy, as separate injections for pramlintide and insulin must be administered. Although marketed insulin formulations generally demonstrate a peak action in 60-90 minutes, pramlintide elicits its peak concentration at around 20-30 minutes after administration. Thus, owing to the significant differences in pharmacokinetics of exogenously administered pramlintide and insulin, the therapy fails to elicit its action otherwise produced by the endogenous hormones. Hence, strategies such as delaying the release of pramlintide by using inorganic polymers like silica, synthetic polymers like polycaprolactone, and lipids have been employed. Also, approaches like noncovalent conjugation, polyelectrolyte complexation, and use of amphiphilic excipients for codelivery of insulin and pramlintide have been explored to address the issues with pramlintide delivery and improve patient adherence to the therapy. This approach may usher in a new era of diabetes management, offering patients multiple options to tailor their treatment and improve their quality of life. SIGNIFICANCE STATEMENT: To our knowledge, this is the first report that summarizes various challenges in insulin and pramlintide codelivery and strategies to overcome them. The paper also provides deeper insights into various novel formulation strategies for pramlintide that could further broaden the reader's understanding of peptide codelivery.
Collapse
Affiliation(s)
- Sai Pradyuth Kommera
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, India
| | - Ankur Kumar
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, India
| |
Collapse
|
8
|
Breunig S, Quijano JC, Donohue C, Henrickson A, Demeler B, Ku HT, Tirrell DA. Incorporation of Aliphatic Proline Residues into Recombinantly Produced Insulin. ACS Chem Biol 2023; 18:2574-2581. [PMID: 37960878 PMCID: PMC10728891 DOI: 10.1021/acschembio.3c00561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023]
Abstract
Analogs of proline can be used to expand the chemical space about the residue while maintaining its uniquely restricted conformational space. Here, we demonstrate the incorporation of 4R-methylproline, 4S-methylproline, and 4-methyleneproline into recombinant insulin expressed in Escherichia coli. These modified proline residues, introduced at position B28, change the biophysical properties of insulin: Incorporation of 4-methyleneproline at B28 accelerates fibril formation, while 4-methylation speeds dissociation from the pharmaceutically formulated hexamer. This work expands the scope of proline analogs amenable to incorporation into recombinant proteins and demonstrates how noncanonical amino acid mutagenesis can be used to engineer the therapeutically relevant properties of protein drugs.
Collapse
Affiliation(s)
- Stephanie
L. Breunig
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Janine C. Quijano
- Department
of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Institute City
of Hope, Duarte, California 91010, United States
| | - Cecile Donohue
- Department
of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Institute City
of Hope, Duarte, California 91010, United States
| | - Amy Henrickson
- Department
of Chemistry and Biochemistry, University
of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - Borries Demeler
- Department
of Chemistry and Biochemistry, University
of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
- Department
of Chemistry and Biochemistry, University
of Montana, Missoula, Montana 59801, United States
| | - Hsun Teresa Ku
- Department
of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Institute City
of Hope, Duarte, California 91010, United States
- Irell &
Manella Graduate School of Biological Science, City of Hope, Duarte, California 91010, United
States
| | - David A. Tirrell
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
9
|
Niloy KK, Lowe TL. Injectable systems for long-lasting insulin therapy. Adv Drug Deliv Rev 2023; 203:115121. [PMID: 37898336 DOI: 10.1016/j.addr.2023.115121] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
Insulin therapy is the mainstay to treat diabetes characterizedd by hyperglycemia. However, its short half-life of only 4-6 min limits its effectiveness in treating chronic diabetes. Advances in recombinant DNA technology and protein engineering have led to several insulin analogue products that have up to 42 h of glycemic control. However, these insulin analogues still require once- or twice-daily injections for optimal glycemic control and have poor patient compliance and adherence issues. To achieve insulin release for more than one day, different injectable delivery systems including microspheres, in situ forming depots, nanoparticles and composite systems have been developed. Several of these delivery systems have advanced to clinical trials for once-weekly insulin injection. This review comprehensively summarizes the developments of injectable insulin analogs and delivery systems covering the whole field of injectable long-lasting insulin technologies from prototype design, preclinical studies, clinical trials to marketed products for the treatment of diabetes.
Collapse
Affiliation(s)
- Kumar Kulldeep Niloy
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Tao L Lowe
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
10
|
Bohr F, Bohr SSR, Mishra NK, González-Foutel NS, Pinholt HD, Wu S, Nielsen EM, Zhang M, Kjaergaard M, Jensen KJ, Hatzakis NS. Enhanced hexamerization of insulin via assembly pathway rerouting revealed by single particle studies. Commun Biol 2023; 6:178. [PMID: 36792809 PMCID: PMC9932072 DOI: 10.1038/s42003-022-04386-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 12/20/2022] [Indexed: 02/17/2023] Open
Abstract
Insulin formulations with diverse oligomerization states are the hallmark of interventions for the treatment of diabetes. Here using single-molecule recordings we firstly reveal that insulin oligomerization can operate via monomeric additions and secondly quantify the existence, abundance and kinetic characterization of diverse insulin assembly and disassembly pathways involving addition of monomeric, dimeric or tetrameric insulin species. We propose and experimentally validate a model where the insulin self-assembly pathway is rerouted, favoring monomeric or oligomeric assembly, by solution concentration, additives and formulations. Combining our practically complete kinetic characterization with rate simulations, we calculate the abundance of each oligomeric species from nM to mM offering mechanistic insights and the relative abundance of all oligomeric forms at concentrations relevant both for secreted and administrated insulin. These reveal a high abundance of all oligomers and a significant fraction of hexamer resulting in practically halved bioavailable monomer concentration. In addition to providing fundamental new insights, the results and toolbox presented here can be universally applied, contributing to the development of optimal insulin formulations and the deciphering of oligomerization mechanisms for additional proteins.
Collapse
Affiliation(s)
- Freja Bohr
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren S-R Bohr
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Narendra Kumar Mishra
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
| | - Nicolás Sebastian González-Foutel
- Department of Molecular Biology and Genetics, The Danish Research Institute for Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, and Center for Proteins in Memory PROMEMO, Danish National Research Foundation, Aarhus University, Aarhus, Denmark
| | - Henrik Dahl Pinholt
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Physics Department, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shunliang Wu
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Milan Nielsen
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Min Zhang
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Magnus Kjaergaard
- Department of Molecular Biology and Genetics, The Danish Research Institute for Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, and Center for Proteins in Memory PROMEMO, Danish National Research Foundation, Aarhus University, Aarhus, Denmark
| | - Knud J Jensen
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark.
| | - Nikos S Hatzakis
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
11
|
Thalhammer A, Bröker NK. Biophysical Approaches for the Characterization of Protein-Metabolite Interactions. Methods Mol Biol 2023; 2554:199-229. [PMID: 36178628 DOI: 10.1007/978-1-0716-2624-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
With an estimate of hundred thousands of protein molecules per cell and the number of metabolites several orders of magnitude higher, protein-metabolite interactions are omnipresent. In vitro analyses are one of the main pillars on the way to establish a solid understanding of how these interactions contribute to maintaining cellular homeostasis. A repertoire of biophysical techniques is available by which protein-metabolite interactions can be quantitatively characterized in terms of affinity, specificity, and kinetics in a broad variety of solution environments. Several of those provide information on local or global conformational changes of the protein partner in response to ligand binding. This review chapter gives an overview of the state-of-the-art biophysical toolbox for the study of protein-metabolite interactions. It briefly introduces basic principles, highlights recent examples from the literature, and pinpoints promising future directions.
Collapse
Affiliation(s)
- Anja Thalhammer
- Physical Biochemistry, University of Potsdam, Potsdam, Germany.
| | - Nina K Bröker
- Physical Biochemistry, University of Potsdam, Potsdam, Germany
- Health and Medical University Potsdam, Potsdam, Germany
| |
Collapse
|
12
|
Bramlage P, Tittel SR, Müther S, Reinhart-Steininger B, Haberland H, Khodaverdi S, Zimny S, Ohlenschläger U, Lanzinger S, Haak T. A comparison of the rapid-acting insulin analogue glulisine with lispro and aspart for the pump treatment of patients with type 1 diabetes. Acta Diabetol 2022; 59:1453-1460. [PMID: 35933650 DOI: 10.1007/s00592-022-01939-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/03/2022] [Indexed: 11/29/2022]
Abstract
AIMS (1) To describe the population of patients with type 1 diabetes (T1DM) using the rapid-acting insulin analogue glulisine versus lispro and aspart during continuous subcutaneous insulin infusion (CSII); (2) to describe insulin relative effectiveness based on hemoglobin A1c (HbA1c), fasting blood glucose (FBG) and dose; (3) to determine rates of hyperglycemia, hypoglycemia, and diabetic ketoacidosis (DKA). METHODS The analysis used March 2021 data from the Diabetes-Patienten-Verlaufsdokumentation registry, which contains data of 618,903 patients with diabetes. Patients were propensity-matched by age, sex, and diabetes duration. RESULTS Overall, 42,736 patients of any age were eligible for analysis based on insulin pump usage with either glulisine (N = 707) or lispro/aspart (N = 42,029) between 2004 and 2020. Patients receiving glulisine were older (median 20.0 vs. 16.2 years), equally often male (47.2% vs. 47.8%) and had a longer diabetes duration (median 9.4 vs. 7.4 years). After propensity score matching, 707 pairs remained (total N = 1414). Patient characteristics between groups were similar. Achieved HbA1c values were also comparable: 8.04%, 64 mmol/mol versus 7.96%, 63 mmol/mol for glulisine and lispro/aspart [LS mean difference 0.08 (95%CI - 0.08, 0.25)]. FBG was 9.37 mmol/L (168.9 mg/dL) and 9.58 mmol/L (172.6 mg/dL) in the glulisine and lispro/aspart groups [LS mean diff. - 0.21; (95%CI - 1.13, 0.72)]. Total daily insulin doses and prandial to total insulin ratios were also similar. Glulisine group patients had higher rates of lipodystrophy (0.85% vs. 0.71%) (LS mean diff. 0.18 [95% CI - 1.01, 1.38]) and non-severe DKA (3.11% vs. 0.57%; p = 0.002). Fewer patients in the glulisine group had severe hypoglycemic events (7.66 vs. 9.09; p = 0.333) and severe ketoacidosis events (0.57% vs. 1.56%; p = 0.082) but more had hypoglycemic coma events (p = 0.773), although the differences were not statistically significant. CONCLUSIONS Insulin glulisine had comparable glucose control to lispro/aspart. The use of glulisine was less frequent in the present analysis compared to the previous trials.
Collapse
Affiliation(s)
- Peter Bramlage
- Institut für Pharmakologie und Präventive Medizin, Bahnhofstrasse 20, 49661, Cloppenburg, Germany.
| | - Sascha R Tittel
- Institut für Epidemiologie und Medizinische Biometrie, ZIBMT, Universität Ulm, Ulm, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Silvia Müther
- Diabetes Zentrum für Kinder und Jugendliche, DRK Kliniken Berlin, Berlin, Germany
| | - Birgit Reinhart-Steininger
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Holger Haberland
- Kinderendokrinologische Ambulanz, Diabeteszentrum für Kinder und Jugendliche, Sozialpädiatrisches Zentrum, Sana Kliniken Berlin-Brandenburg GmbH, Berlin, Germany
| | | | | | | | - Stefanie Lanzinger
- Institut für Epidemiologie und Medizinische Biometrie, ZIBMT, Universität Ulm, Ulm, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Haak
- Diabetes Zentrum Mergentheim, Bad Mergentheim, Germany
| |
Collapse
|
13
|
Teigen IA, Riaz M, Åm MK, Christiansen SC, Carlsen SM. Vasodilatory effects of glucagon: A possible new approach to enhanced subcutaneous insulin absorption in artificial pancreas devices. Front Bioeng Biotechnol 2022; 10:986858. [PMID: 36213069 PMCID: PMC9532737 DOI: 10.3389/fbioe.2022.986858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Patients with diabetes mellitus type 1 depend on exogenous insulin to keep their blood glucose concentrations within the desired range. Subcutaneous bihormonal artificial pancreas devices that can measure glucose concentrations continuously and autonomously calculate and deliver insulin and glucagon infusions is a promising new treatment option for these patients. The slow absorption rate of insulin from subcutaneous tissue is perhaps the most important factor preventing the development of a fully automated artificial pancreas using subcutaneous insulin delivery. Subcutaneous insulin absorption is influenced by several factors, among which local subcutaneous blood flow is one of the most prominent. We have discovered that micro-doses of glucagon may cause a substantial increase in local subcutaneous blood flow. This paper discusses how the local vasodilative effects of micro-doses of glucagon might be utilised to improve the performance of subcutaneous bihormonal artificial pancreas devices. We map out the early stages of our hypothesis as a disruptive novel approach, where we propose to use glucagon as a vasodilator to accelerate the absorption of meal boluses of insulin, besides using it conventionally to treat hypoglycaemia.
Collapse
Affiliation(s)
- Ingrid Anna Teigen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Misbah Riaz
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Marte Kierulf Åm
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sverre Christian Christiansen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Sven Magnus Carlsen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
14
|
Maikawa CL, Nguyen LT, Mann JL, Appel EA. Formulation Excipients and Their Role in Insulin Stability and Association State in Formulation. Pharm Res 2022; 39:2721-2728. [PMID: 35978148 PMCID: PMC9633423 DOI: 10.1007/s11095-022-03367-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
While excipients are often overlooked as the "inactive" ingredients in pharmaceutical formulations, they often play a critical role in protein stability and absorption kinetics. Recent work has identified an ultrafast absorbing insulin formulation that is the result of excipient modifications. Specifically, the insulin monomer can be isolated by replacing zinc and the phenolic preservative metacresol with phenoxyethanol as an antimicrobial agent and an amphiphilic acrylamide copolymer excipient for stability. A greater understanding is needed of the interplay between excipients, insulin association state, and stability in order to optimize this formulation. Here, we formulated insulin with different preservatives and stabilizing excipient concentrations using both insulin lispro and regular human insulin and assessed the insulin association states using analytical ultracentrifugation as well as formulation stability. We determined that phenoxyethanol is required to eliminate hexamers and promote a high monomer content even in a zinc-free lispro formulation. There is also a concentration dependent relationship between the concentration of polyacrylamide-based copolymer excipient and insulin stability, where a concentration greater than 0.1 g/mL copolymer is required for a mostly monomeric zinc-free lispro formulation to achieve stability exceeding that of Humalog in a stressed aging assay. Further, we determined that under the formulation conditions tested zinc-free regular human insulin remains primarily hexameric and is not at this time a promising candidate for rapid-acting formulations.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, 94305, USA
| | - Leslee T Nguyen
- Department of Biochemistry, Stanford University, Stanford, 94305, USA
| | - Joseph L Mann
- Department of Materials Science & Engineering, Stanford University, Stanford, 94305, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, 94305, USA. .,Department of Materials Science & Engineering, Stanford University, Stanford, 94305, USA. .,Department of Pediatrics (Endocrinology), Stanford University, Stanford, 94305, USA. .,ChEM-H Institute, Stanford University, Stanford, CA, 94305, USA. .,Woods Institute for the Environment, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
15
|
Khadria A, Paavola CD, Maslov K, Valenzuela FA, Sperry AE, Cox AL, Cao R, Shi J, Brown-Augsburger PL, Lozano E, Blankenship RL, Majumdar R, Bradley SA, Beals JM, Oladipupo SS, Wang LV. Photoacoustic imaging reveals mechanisms of rapid-acting insulin formulations dynamics at the injection site. Mol Metab 2022; 62:101522. [PMID: 35671972 PMCID: PMC9207296 DOI: 10.1016/j.molmet.2022.101522] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE Ultra-rapid insulin formulations control postprandial hyperglycemia; however, inadequate understanding of injection site absorption mechanisms is limiting further advancement. We used photoacoustic imaging to investigate the injection site dynamics of dye-labeled insulin lispro in the Humalog® and Lyumjev® formulations using the murine ear cutaneous model and correlated it with results from unlabeled insulin lispro in pig subcutaneous injection model. METHODS We employed dual-wavelength optical-resolution photoacoustic microscopy to study the absorption and diffusion of the near-infrared dye-labeled insulin lispro in the Humalog and Lyumjev formulations in mouse ears. We mathematically modeled the experimental data to calculate the absorption rate constants and diffusion coefficients. We studied the pharmacokinetics of the unlabeled insulin lispro in both the Humalog and Lyumjev formulations as well as a formulation lacking both the zinc and phenolic preservative in pigs. The association state of insulin lispro in each of the formulations was characterized using SV-AUC and NMR spectroscopy. RESULTS Through experiments using murine and swine models, we show that the hexamer dissociation rate of insulin lispro is not the absorption rate-limiting step. We demonstrated that the excipients in the Lyumjev formulation produce local tissue expansion and speed both insulin diffusion and microvascular absorption. We also show that the diffusion of insulin lispro at the injection site drives its initial absorption; however, the rate at which the insulin lispro crosses the blood vessels is its overall absorption rate-limiting step. CONCLUSIONS This study provides insights into injection site dynamics of insulin lispro and the impact of formulation excipients. It also demonstrates photoacoustic microscopy as a promising tool for studying protein therapeutics. The results from this study address critical questions around the subcutaneous behavior of insulin lispro and the formulation excipients, which could be useful to make faster and better controlled insulin formulations in the future.
Collapse
Affiliation(s)
- Anjul Khadria
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Chad D Paavola
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Konstantin Maslov
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Francisco A Valenzuela
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Andrea E Sperry
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Amy L Cox
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Rui Cao
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Junhui Shi
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | | | - Emmanuel Lozano
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Ross L Blankenship
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Ranajoy Majumdar
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Scott A Bradley
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - John M Beals
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Biotechnology Center, San Diego, CA, 92121, USA.
| | - Sunday S Oladipupo
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA.
| | - Lihong V Wang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA; Caltech Optical Imaging Laboratory, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
16
|
Abdelwaly EA, Mohamed AA, El-Kosasy AM, Ayad MF. Simultaneous quantitative determination of insulin aspart and insulin degludec in human plasma using simple shoot LC method. Biomed Chromatogr 2021; 36:e5292. [PMID: 34854111 DOI: 10.1002/bmc.5292] [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: 05/25/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 11/08/2022]
Abstract
Combining short-acting and long-acting insulin analogs was a real challenge that was overcome by NovoNordisk through the co-formulation of insulin aspart and insulin degludec in single-dosage form. The proposed study provides a simple, short, and reliable HPLC method with diode array detection that is developed and validated for the simultaneous determination of insulin aspart and insulin degludec in human plasma. The proposed method achieved good separation between the two analytes utilizing a C8 column at 35°C in a very short run time (6 min), with a simple, low-cost, and reliable extraction method through precipitation of plasma protein. Gradient elution was applied using a mobile phase consisting of 0.1 M sodium sulfate (pH 3.4) and acetonitrile. The method was validated according to EMA Guideline on Bioanalytical Validation. The proposed method had a linear range from 3.0 to 300 μg/mL for insulin aspart and from 3.5 to 300 μg/mL for insulin degludec. The intra- and inter-day precision of insulin aspart were 0.36-3.33% and 1.59-8.84%, respectively, and accuracy was between 10.06 and 3.09% The intra- and inter-day precision of insulin degludec were 0.29-1.93% and 0.89-5.14%, respectively, and accuracy was between -5.29 and 3.91%.
Collapse
Affiliation(s)
| | - Abeer A Mohamed
- Egyptian Drug Authority, Cairo, Egypt.,Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Amira M El-Kosasy
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Miriam F Ayad
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
17
|
Maikawa CL, Chen PC, Vuong ET, Nguyen LT, Mann JL, d'Aquino AI, Lal RA, Maahs DM, Buckingham BA, Appel EA. Ultra-Fast Insulin-Pramlintide Co-Formulation for Improved Glucose Management in Diabetic Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101575. [PMID: 34499434 PMCID: PMC8564421 DOI: 10.1002/advs.202101575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/07/2021] [Indexed: 05/04/2023]
Abstract
Dual-hormone replacement therapy with insulin and amylin in patients with type 1 diabetes has the potential to improve glucose management. Unfortunately, currently available formulations require burdensome separate injections at mealtimes and have disparate pharmacokinetics that do not mimic endogenous co-secretion. Here, amphiphilic acrylamide copolymers are used to create a stable co-formulation of monomeric insulin and amylin analogues (lispro and pramlintide) with synchronous pharmacokinetics and ultra-rapid action. The co-formulation is stable for over 16 h under stressed aging conditions, whereas commercial insulin lispro (Humalog) aggregates in 8 h. The faster pharmacokinetics of monomeric insulin in this co-formulation result in increased insulin-pramlintide overlap of 75 ± 6% compared to only 47 ± 7% for separate injections. The co-formulation results in similar delay in gastric emptying compared to pramlintide delivered separately. In a glucose challenge, in rats, the co-formulation reduces deviation from baseline glucose compared to insulin only, or separate insulin and pramlintide administrations. Further, comparison of interspecies pharmacokinetics of monomeric pramlintide suggests that pharmacokinetics observed for the co-formulation will be well preserved in future translation to humans. Together these results suggest that the co-formulation has the potential to improve mealtime glucose management and reduce patient burden in the treatment of diabetes.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Peyton C Chen
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Eric T Vuong
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Leslee T Nguyen
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Joseph L Mann
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Andrea I d'Aquino
- Department of Materials Science & 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
| | - David M Maahs
- 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 & 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
|
18
|
Mann JL, Maikawa CL, Smith AAA, Grosskopf AK, Baker SW, Roth GA, Meis CM, Gale EC, Liong CS, Correa S, Chan D, Stapleton LM, Yu AC, Muir B, Howard S, Postma A, Appel EA. An ultrafast insulin formulation enabled by high-throughput screening of engineered polymeric excipients. Sci Transl Med 2021; 12:12/550/eaba6676. [PMID: 32611683 DOI: 10.1126/scitranslmed.aba6676] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
Insulin has been used to treat diabetes for almost 100 years; yet, current rapid-acting insulin formulations do not have sufficiently fast pharmacokinetics to maintain tight glycemic control at mealtimes. Dissociation of the insulin hexamer, the primary association state of insulin in rapid-acting formulations, is the rate-limiting step that leads to delayed onset and extended duration of action. A formulation of insulin monomers would more closely mimic endogenous postprandial insulin secretion, but monomeric insulin is unstable in solution using present formulation strategies and rapidly aggregates into amyloid fibrils. Here, we implement high-throughput-controlled radical polymerization techniques to generate a large library of acrylamide carrier/dopant copolymer (AC/DC) excipients designed to reduce insulin aggregation. Our top-performing AC/DC excipient candidate enabled the development of an ultrafast-absorbing insulin lispro (UFAL) formulation, which remains stable under stressed aging conditions for 25 ± 1 hours compared to 5 ± 2 hours for commercial fast-acting insulin lispro formulations (Humalog). In a porcine model of insulin-deficient diabetes, UFAL exhibited peak action at 9 ± 4 min, whereas commercial Humalog exhibited peak action at 25 ± 10 min. These ultrafast kinetics make UFAL a promising candidate for improving glucose control and reducing burden for patients with diabetes.
Collapse
Affiliation(s)
- Joseph L Mann
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Anton A A Smith
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA.,Department of Science and Technology, Aarhus University, 8000 Aarhus, Denmark
| | - Abigail K Grosskopf
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Sam W Baker
- Department of Comparative Medicine, Stanford University, Palo Alto, CA 94305, USA
| | - Gillie A Roth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Catherine M Meis
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Emily C Gale
- Department of Biochemistry, Stanford University, Palo Alto, CA 94305, USA
| | - Celine S Liong
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Santiago Correa
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Doreen Chan
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | | | - Anthony C Yu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Ben Muir
- CSIRO Manufacturing, Clayton, VIC 3168, Australia
| | - Shaun Howard
- CSIRO Manufacturing, Clayton, VIC 3168, Australia
| | - Almar Postma
- CSIRO Manufacturing, Clayton, VIC 3168, Australia
| | - Eric A Appel
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA. .,Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.,ChEM-H Institute, Stanford University, Stanford, CA 94305, USA.,Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
19
|
Mulka A, Lewis BE, Mao L, Sharafieh R, Kesserwan S, Wu R, Kreutzer DL, Klueh U. Phenolic Preservative Removal from Commercial Insulin Formulations Reduces Tissue Inflammation while Maintaining Euglycemia. ACS Pharmacol Transl Sci 2021; 4:1161-1174. [PMID: 34151206 DOI: 10.1021/acsptsci.1c00047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Indexed: 11/28/2022]
Abstract
Background: Exogenous insulin therapy requires stabilization of the insulin molecule, which is achieved through the use of excipients (e.g., phenolic preservatives (PP)) that provide protein stability, sterility and prolong insulin shelf life. However, our laboratory recently reported that PP, (e.g., m-creosol and phenol) are also cytotoxic, inducing inflammation and fibrosis. Optimizing PP levels through filtration would balance the need for insulin preservation with PP-induced inflammation. Method: Zeolite Y (Z-Y), a size-exclusion-based resin, was employed to remove PP from commercial insulin formulations (Humalog) before infusion. Results: PP removal significantly decreased cell toxicity in vitro and inflammation in vivo. Infusion site histological analysis after a 3 day study demonstrated that leukocyte accumulation increased with nonfiltered preparations but decreased after filtration. Additional studies demonstrated that a Z-Y fabricated filter effectively removed excess PP such that the filtered insulin solution achieved equivalent glycemic control in diabetic mice when compared to nonfiltered insulin. Conclusion: This approach represents the proof of concept that using Z-Y for in-line PP removal assists in lowering inflammation at the site of insulin infusion and thus could lead to extending the functional lifespan of insulin infusion sets in vivo.
Collapse
Affiliation(s)
- Adam Mulka
- Department of Biomedical Engineering, Integrative Biosciences Center, Wayne State University, Detroit, Michigan 48202,United States
| | - Brianne E Lewis
- Department of Biomedical Engineering, Integrative Biosciences Center, Wayne State University, Detroit, Michigan 48202,United States
| | - Li Mao
- Department of Biomedical Engineering, Integrative Biosciences Center, Wayne State University, Detroit, Michigan 48202,United States
| | - Roshanak Sharafieh
- Department of Surgery, School of Medicine, University of Connecticut, Farmington, Connecticut 06030-2100, United States
| | - Shereen Kesserwan
- Department of Biomedical Engineering, Integrative Biosciences Center, Wayne State University, Detroit, Michigan 48202,United States
| | - Rong Wu
- Connecticut Convergence Institute, School of Medicine, University of Connecticut, Farmington, Connecticut 06030-6022, United States
| | - Donald L Kreutzer
- Department of Surgery, School of Medicine, University of Connecticut, Farmington, Connecticut 06030-2100, United States
| | - Ulrike Klueh
- Department of Biomedical Engineering, Integrative Biosciences Center, Wayne State University, Detroit, Michigan 48202,United States
| |
Collapse
|
20
|
Maikawa CL, d'Aquino AI, Lal RA, Buckingham BA, Appel EA. Engineering biopharmaceutical formulations to improve diabetes management. Sci Transl Med 2021; 13:eabd6726. [PMID: 33504649 PMCID: PMC8004356 DOI: 10.1126/scitranslmed.abd6726] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
Insulin was first isolated almost a century ago, yet commercial formulations of insulin and its analogs for hormone replacement therapy still fall short of appropriately mimicking endogenous glycemic control. Moreover, the controlled delivery of complementary hormones (such as amylin or glucagon) is complicated by instability of the pharmacologic agents and complexity of maintaining multiple infusions. In this review, we highlight the advantages and limitations of recent advances in drug formulation that improve protein stability and pharmacokinetics, prolong drug delivery, or enable alternative dosage forms for the management of diabetes. With controlled delivery, these formulations could improve closed-loop glycemic control.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Andrea I d'Aquino
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Rayhan A Lal
- Department of Medicine (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Bruce A Buckingham
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Gillis RB, Solomon HV, Govada L, Oldham NJ, Dinu V, Jiwani SI, Gyasi-Antwi P, Coffey F, Meal A, Morgan PS, Harding SE, Helliwell JR, Chayen NE, Adams GG. Analysis of insulin glulisine at the molecular level by X-ray crystallography and biophysical techniques. Sci Rep 2021; 11:1737. [PMID: 33462295 PMCID: PMC7814034 DOI: 10.1038/s41598-021-81251-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 12/09/2020] [Indexed: 11/16/2022] Open
Abstract
This study concerns glulisine, a rapid-acting insulin analogue that plays a fundamental role in diabetes management. We have applied a combination of methods namely X-ray crystallography, and biophysical characterisation to provide a detailed insight into the structure and function of glulisine. X-ray data provided structural information to a resolution of 1.26 Å. Crystals belonged to the H3 space group with hexagonal (centred trigonal) cell dimensions a = b = 82.44 and c = 33.65 Å with two molecules in the asymmetric unit. A unique position of D21Glu, not present in other fast-acting analogues, pointing inwards rather than to the outside surface was observed. This reduces interactions with neighbouring molecules thereby increasing preference of the dimer form. Sedimentation velocity/equilibrium studies revealed a trinary system of dimers and hexamers/dihexamers in dynamic equilibrium. This new information may lead to better understanding of the pharmacokinetic and pharmacodynamic behaviour of glulisine which might aid in improving formulation regarding its fast-acting role and reducing side effects of this drug.
Collapse
Affiliation(s)
- Richard B Gillis
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2HA, UK.
| | - Hodaya V Solomon
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK
| | - Lata Govada
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK
| | - Neil J Oldham
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Vlad Dinu
- National Centre for Macromolecular Hydrodynamics, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| | - Shahwar Imran Jiwani
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2HA, UK
| | - Philemon Gyasi-Antwi
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2HA, UK
| | - Frank Coffey
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2HA, UK
| | - Andy Meal
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2HA, UK
| | - Paul S Morgan
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2HA, UK
| | - Stephen E Harding
- National Centre for Macromolecular Hydrodynamics, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK.,Universitetet I Oslo, St. Olavs plass, Postboks 6762, 0130, Oslo, Norway
| | - John R Helliwell
- Department of Chemistry, University of Manchester, Manchester, M13 9PL, UK
| | - Naomi E Chayen
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK.
| | - Gary G Adams
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2HA, UK.
| |
Collapse
|
22
|
Beji O, Gillis RB, Dinu V, Jiwani SI, Gyasi-Antwi P, Fisk ID, Meal A, Morgan PS, Harding SE, Huang S, Agugini G, Fedele F, Adams GG. Exploration of temperature and shelf-life dependency of the therapeutically available Insulin Detemir. Eur J Pharm Biopharm 2020; 152:340-347. [PMID: 32446962 DOI: 10.1016/j.ejpb.2020.05.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 05/11/2020] [Accepted: 05/17/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE Insulin, in typical use, undergoes multiple changes in temperature; from refrigerator, to room temperature, to body temperature. Although long-term storage temperature has been well-studied, the short term changes to insulin are yet to be determined. Insulin detemir (IDet) is a clinically available, slow-acting, synthetic analogue characterised by the conjugation of a C14 fatty acid. The function of this modification is to cause the insulin to form multi-hexameric species, thus retarding the pharmacokinetic rate of action. In this investigation, the temperature dependence properties of this synthetic analogue is probed, as well as expiration. METHODS Dynamic light scattering (DLS) and viscometry were employed to assess the effect of temperature upon IDet. Mass spectrometry was also used to probe the impact of shelf-life and the presence of certain excipients. RESULTS IDet was compared with eight other insulins, including human recombinant, three fast-acting analogues and two other slow-acting analogues. Of all nine insulins, IDet was the only analogue to show temperature dependent behaviour, between 20 °C and 37 °C, when probed with non-invasive backscatter dynamic light scattering. Upon further investigation, IDet observed significant changes in size related to temperature, direction of temperature (heated/cooled) and expiration with cross-correlation observed amongst all 4 parameters. CONCLUSIONS These findings are critical to our understanding of the behaviour of this particular clinically relevant drug, as it will allow the development of future generations of peptide-based therapies with greater clinical efficacy.
Collapse
Affiliation(s)
- Oritsegidenene Beji
- University of Nottingham, School of Biosciences, Sutton Bonington Campus, Leicestershire, UK
| | - Richard B Gillis
- University of Nottingham, School of Health Sciences, Faculty of Medicine and Health Sciences, Queens Medical Centre, Clifton Boulevard, Nottingham, UK.
| | - Vlad Dinu
- University of Nottingham, School of Biosciences, Sutton Bonington Campus, Leicestershire, UK; University of Nottingham, National Centre for Macromolecular Hydrodynamics, Sutton Bonington Campus, Leicestershire, UK
| | - Shahwar I Jiwani
- University of Nottingham, School of Health Sciences, Faculty of Medicine and Health Sciences, Queens Medical Centre, Clifton Boulevard, Nottingham, UK
| | - Philemon Gyasi-Antwi
- University of Nottingham, School of Health Sciences, Faculty of Medicine and Health Sciences, Queens Medical Centre, Clifton Boulevard, Nottingham, UK
| | - Ian D Fisk
- University of Nottingham, School of Biosciences, Sutton Bonington Campus, Leicestershire, UK
| | - Andrew Meal
- University of Nottingham, School of Health Sciences, Faculty of Medicine and Health Sciences, Queens Medical Centre, Clifton Boulevard, Nottingham, UK
| | - Paul S Morgan
- University of Nottingham, School of Medicine, Faculty of Medicine and Health Sciences, Queens Medical Centre, Clifton Boulevard, Nottingham, UK
| | - Stephen E Harding
- University of Nottingham, School of Biosciences, Sutton Bonington Campus, Leicestershire, UK; University of Nottingham, National Centre for Macromolecular Hydrodynamics, Sutton Bonington Campus, Leicestershire, UK; Universitetet I Oslo, Postboks 6762, St. Olavs plass, 0130 Oslo, Norway
| | - Sha Huang
- University of Nottingham, School of Biosciences, Sutton Bonington Campus, Leicestershire, UK
| | - Giulia Agugini
- University of Pavia, Department of Drug Sciences, Pavia, Italy
| | - Federica Fedele
- University of Salento, Piazzetta Tancredi 7, 73100 Lecce, Italy
| | - Gary G Adams
- University of Nottingham, School of Health Sciences, Faculty of Medicine and Health Sciences, Queens Medical Centre, Clifton Boulevard, Nottingham, UK.
| |
Collapse
|
23
|
Frost TS, Jiang L, Zohar Y. Pharmacokinetic Analysis of Epithelial/Endothelial Cell Barriers in Microfluidic Bilayer Devices with an Air-Liquid Interface. MICROMACHINES 2020; 11:mi11050536. [PMID: 32466113 PMCID: PMC7281310 DOI: 10.3390/mi11050536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 12/23/2022]
Abstract
As the range of applications of organs-on-chips is broadening, the evaluation of aerosol-based therapies using a lung-on-a-chip model has become an attractive approach. Inhalation therapies are not only minimally invasive but also provide optimal pharmacokinetic conditions for drug absorption. As drug development evolves, it is likely that better screening through use of organs-on-chips can significantly save time and cost. In this work, bio-aerosols of various compounds including insulin were generated using a jet nebulizer. The aerosol flows were driven through microfluidic bilayer devices establishing an air–liquid interface to mimic the blood–air barrier in human small airways. The aerosol flow in the microfluidic devices has been characterized and adjusted to closely match physiological values. The permeability of several compounds, including paracellular and transcellular biomarkers, across epithelial/endothelial cell barriers was measured. Concentration–time plots were established in microfluidic devices with and without cells; the curves were then utilized to extract standard pharmacokinetic parameters such as the area under the curve, maximum concentration, and time to maximum concentration. The cell barrier significantly affected the measured pharmacokinetic parameters, as compound absorption through the barrier decreases with its increasing molecular size. Aerosolizing insulin can lead to the formation of fibrils, prior to its entry to the microfluidic device, with a substantially larger apparent molecular size effectively blocking its paracellular transport. The results demonstrate the advantage of using lung-on-a-chip for drug discovery with applications such as development of novel inhaled therapies.
Collapse
Affiliation(s)
- Timothy S. Frost
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA;
- Correspondence:
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| | - Yitshak Zohar
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA;
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| |
Collapse
|
24
|
Maikawa CL, Smith AAA, Zou L, Roth GA, Gale EC, Stapleton LM, Baker SW, Mann JL, Yu AC, Correa S, Grosskopf AK, Liong CS, Meis CM, Chan D, Troxell M, Maahs DM, Buckingham BA, Webber MJ, Appel EA. A co-formulation of supramolecularly stabilized insulin and pramlintide enhances mealtime glucagon suppression in diabetic pigs. Nat Biomed Eng 2020; 4:507-517. [PMID: 32393892 PMCID: PMC7274092 DOI: 10.1038/s41551-020-0555-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
Treatment of patients with diabetes with insulin and pramlintide (an amylin analogue) is more effective than treatment with insulin only. However, because mixtures of insulin and pramlintide are unstable and have to be injected separately, amylin analogues are only used by 1.5% of people with diabetes needing rapid-acting insulin. Here, we show that the supramolecular modification of insulin and pramlintide with cucurbit[7]uril-conjugated polyethylene glycol improves the pharmacokinetics of the dual-hormone therapy and enhances postprandial glucagon suppression in diabetic pigs. The co-formulation is stable for over 100 h at 37 °C under continuous agitation, whereas commercial formulations of insulin analogues aggregate after 10 h under similar conditions. In diabetic rats, the administration of the stabilized co-formulation increased the area-of-overlap ratio of the pharmacokinetic curves of pramlintide and insulin from 0.4 ± 0.2 to 0.7 ± 0.1 (mean ± s.d.) for the separate administration of the hormones. The co-administration of supramolecularly stabilized insulin and pramlintide better mimics the endogenous kinetics of co-secreted insulin and amylin, and holds promise as a dual-hormone replacement therapy.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Anton A A Smith
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Department of Science and Technology, Aarhus University, Aarhus, Denmark
| | - Lei Zou
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Gillie A Roth
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Emily C Gale
- Department of Biochemistry, Stanford University, Stanford, CA, USA
| | | | - Sam W Baker
- Department of Comparative Medicine, Stanford University, Stanford, CA, USA
| | - Joseph L Mann
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Anthony C Yu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Santiago Correa
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | | | - Celine S Liong
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Catherine M Meis
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Doreen Chan
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Megan Troxell
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - David M Maahs
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA
- Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Bruce A Buckingham
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA
- Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Matthew J Webber
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA.
- Diabetes Research Center, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Wolff M, Schüler A, Gast K, Seckler R, Evers A, Pfeiffer-Marek S, Kurz M, Nagel N, Haack T, Wagner M, Thalhammer A. Self-Assembly of Exendin-4-Derived Dual Peptide Agonists is Mediated by Acylation and Correlated to the Length of Conjugated Fatty Acyl Chains. Mol Pharm 2020; 17:965-978. [DOI: 10.1021/acs.molpharmaceut.9b01195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Martin Wolff
- Physical Biochemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, D-14476 Potsdam, Germany
| | - Anja Schüler
- Physical Biochemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, D-14476 Potsdam, Germany
| | - Klaus Gast
- Physical Biochemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, D-14476 Potsdam, Germany
| | - Robert Seckler
- Physical Biochemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, D-14476 Potsdam, Germany
| | - Andreas Evers
- Sanofi-Aventis Deutschland GmbH, Industrial Park Höchst, D-65926 Frankfurt, Germany
| | | | - Michael Kurz
- Sanofi-Aventis Deutschland GmbH, Industrial Park Höchst, D-65926 Frankfurt, Germany
| | - Norbert Nagel
- Sanofi-Aventis Deutschland GmbH, Industrial Park Höchst, D-65926 Frankfurt, Germany
| | - Torsten Haack
- Sanofi-Aventis Deutschland GmbH, Industrial Park Höchst, D-65926 Frankfurt, Germany
| | - Michael Wagner
- Sanofi-Aventis Deutschland GmbH, Industrial Park Höchst, D-65926 Frankfurt, Germany
| | - Anja Thalhammer
- Physical Biochemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, D-14476 Potsdam, Germany
| |
Collapse
|
26
|
Maikawa CL, Smith AAA, Zou L, Meis CM, Mann JL, Webber MJ, Appel EA. Stable Monomeric Insulin Formulations Enabled by Supramolecular PEGylation of Insulin Analogues. ADVANCED THERAPEUTICS 2020; 3:1900094. [PMID: 32190729 PMCID: PMC7079736 DOI: 10.1002/adtp.201900094] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Indexed: 12/17/2022]
Abstract
Current "fast-acting" insulin analogues contain amino acid modifications meant to inhibit dimer formation and shift the equilibrium of association states toward the monomeric state. However, the insulin monomer is highly unstable and current formulation techniques require insulin to primarily exist as hexamers to prevent aggregation into inactive and immunogenic amyloids. Insulin formulation excipients have thus been traditionally selected to promote insulin association into the hexameric form to enhance formulation stability. This study exploits a novel excipient for the supramolecular PEGylation of insulin analogues, including aspart and lispro, to enhance the stability and maximize the prevalence of insulin monomers in formulation. Using multiple techniques, it is demonstrated that judicious choice of formulation excipients (tonicity agents and parenteral preservatives) enables insulin analogue formulations with 70-80% monomer and supramolecular PEGylation imbued stability under stressed aging for over 100 h without altering the insulin association state. Comparatively, commercial "fast-acting" formulations contain less than 1% monomer and remain stable for only 10 h under the same stressed aging conditions. This simple and effective formulation approach shows promise for next-generation ultrafast insulin formulations with a short duration of action that can reduce the risk of post-prandial hypoglycemia in the treatment of diabetes.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Anton A A Smith
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Lei Zou
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Catherine M Meis
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Joseph L Mann
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Matthew J Webber
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
27
|
Shou K, Bremer A, Rindfleisch T, Knox-Brown P, Hirai M, Rekas A, Garvey CJ, Hincha DK, Stadler AM, Thalhammer A. Conformational selection of the intrinsically disordered plant stress protein COR15A in response to solution osmolarity - an X-ray and light scattering study. Phys Chem Chem Phys 2019; 21:18727-18740. [PMID: 31424463 DOI: 10.1039/c9cp01768b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The plant stress protein COR15A stabilizes chloroplast membranes during freezing. COR15A is an intrinsically disordered protein (IDP) in aqueous solution, but acquires an α-helical structure during dehydration or the increase of solution osmolarity. We have used small- and wide-angle X-ray scattering (SAXS/WAXS) combined with static and dynamic light scattering (SLS/DLS) to investigate the structural and hydrodynamic properties of COR15A in response to increasing solution osmolarity. Coarse-grained ensemble modelling allowed a structure-based interpretation of the SAXS data. Our results demonstrate that COR15A behaves as a biomacromolecule with polymer-like properties which strongly depend on solution osmolarity. Biomacromolecular self-assembly occurring at high solvent osmolarity is initiated by the occurrence of two specific structural subpopulations of the COR15A monomer. The osmolarity dependent structural selection mechanism is an elegant way for conformational regulation and assembly of COR15A. It highlights the importance of the polymer-like properties of IDPs for their associated biological function.
Collapse
Affiliation(s)
- Keyun Shou
- Jülich Centre for Neutron Science (JCNS-1) and Institute for Complex Systems (ICS-1), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Falk BT, Liang Y, McCoy MA. Profiling Insulin Oligomeric States by 1H NMR Spectroscopy for Formulation Development of Ultra-Rapid-Acting Insulin. J Pharm Sci 2019; 109:922-926. [PMID: 31449814 DOI: 10.1016/j.xphs.2019.07.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/16/2019] [Accepted: 07/30/2019] [Indexed: 11/15/2022]
Abstract
Formulations that can increase the dissociation of insulin oligomers into monomers/dimers are important considerations in the development of ultra-rapid-acting insulins with faster onset and shorter duration of actions. Here we present a novel strategy to characterize the oligomeric states of insulin in solution that leverages the ability of nuclear magnetic resonance spectroscopy to assess higher-order structure of proteins in solution. The oligomeric structures and solution behaviors of 2 fast-acting insulins, aspart and lispro, with varying excipient concentrations were studied using 1D and diffusion profiling methods. These methods can provide insight on the structural differences and distributions of the molecular association states in different insulin formulations, which is consistent with other orthogonal biophysical characterization tools. In addition, these methods also highlight their sensitivity to subtle changes in solution behaviors in response to excipient that are difficult to monitor with other tools. This work introduces the utility of 1D and diffusion profiling methods to characterize the oligomeric assembly of fast-acting insulins, suggesting promising applications in compound screening, excipient selection, and formulation development of fast-acting insulins as well as other peptide or protein therapeutics.
Collapse
Affiliation(s)
- Bradley T Falk
- Mass Spectrometry and Biophysics, Merck & Co., Inc., Kenilworth, New Jersey 07033
| | - Yingkai Liang
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., West Point, Pennsylvania 19486
| | - Mark A McCoy
- Mass Spectrometry and Biophysics, Merck & Co., Inc., Kenilworth, New Jersey 07033.
| |
Collapse
|
29
|
Nagel N, Graewert MA, Gao M, Heyse W, Jeffries CM, Svergun D, Berchtold H. The quaternary structure of insulin glargine and glulisine under formulation conditions. Biophys Chem 2019; 253:106226. [PMID: 31376619 DOI: 10.1016/j.bpc.2019.106226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/18/2019] [Accepted: 07/10/2019] [Indexed: 11/17/2022]
Abstract
The quaternary structures of insulin glargine and glulisine under formulation conditions and upon dilution using placebo or water were investigated using synchrotron small-angle X-ray scattering. Our results revealed that insulin glulisine in Apidra® is predominantly hexameric in solution with significant fractions of dodecamers and monomers. Upon dilution with placebo, this equilibrium shifts towards monomers. Insulin glargine in Lantus® and Toujeo® is present in a stable hexamer/dimer equilibrium, which is hardly affected by dilution with water down to 1 mg/ml insulin concentration. The results provide exclusive insight into the quaternary structure and thus the association/dissociation properties of the two insulin analogues in marketed formulations.
Collapse
Affiliation(s)
- Norbert Nagel
- Sanofi-Aventis Deutschland GmbH, R&D, Industriepark Höchst, 65926 Frankfurt, Germany.
| | - Melissa A Graewert
- European Molecular Biology Laboratory, Hamburg Unit, c/o DESY, Notkestraße 85, 22603 Hamburg, Germany; BioSAXS GmbH c/o DESY, Notkestraße 85, 22603 Hamburg, Germany
| | - Mimi Gao
- Sanofi-Aventis Deutschland GmbH, R&D, Industriepark Höchst, 65926 Frankfurt, Germany
| | - Winfried Heyse
- Sanofi-Aventis Deutschland GmbH, R&D, Industriepark Höchst, 65926 Frankfurt, Germany
| | - Cy M Jeffries
- European Molecular Biology Laboratory, Hamburg Unit, c/o DESY, Notkestraße 85, 22603 Hamburg, Germany
| | - Dmitri Svergun
- European Molecular Biology Laboratory, Hamburg Unit, c/o DESY, Notkestraße 85, 22603 Hamburg, Germany.
| | - Harald Berchtold
- Sanofi-Aventis Deutschland GmbH, R&D, Industriepark Höchst, 65926 Frankfurt, Germany
| |
Collapse
|
30
|
Sato Y, Mizuno Y, Suganuma K, Shiroto K, Ikeda T, Yamashita K, Kimura T, Yamauchi K, Aizawa T. Pharmacokinetics of insulin disappearance after massive overdosing. Endocr J 2018; 65:1147-1153. [PMID: 30185719 DOI: 10.1507/endocrj.ej18-0118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Long-term glucose supplementation is required to prevent hypoglycemia after massive insulin overdosing. We fitted the blood insulin concentration-time profile to the model: I = A·exp(-a·t) + B·exp(-b·t), where I (μU/mL) is the serum/plasma insulin concentration, A (μU/mL) and B (μU/mL) are the peak insulin concentrations of each component, a (time-1) and b (time-1) are the time constants of each component, and t (h) is the time elapsed from the peak of blood insulin level. Additional components were considered as needed. Patient 1 had auto-injected 600 U NovoRapid® 30Mix, and Patient 2 had auto-injected 300 U Novolet®R (regular) and 1,800 U NovoLet®N (NPH). We used the disappearance of therapeutic doses of the respective insulin in healthy individuals as controls, and we obtained parameters by Excel solver. In Patient 1, the parameter values were A = 1490.04 and a = 0.15 for insulin aspart and B = 60.66 and b = 0.04 for protaminated aspart. In Patient 2, the values were A = 784.45 and a = 0.38 for regular insulin and B = 395.84 and b = 0.03 for NPH. Compared with controls, the half-lives (t1/2) for insulin aspart and protaminated aspart were 4 and 2 times longer, respectively, in Patient 1. In Patient 2, the t1/2 for regular and NPH insulin were 2 and 7 times longer than those in the controls, respectively. In conclusion, the t1/2 for insulin was elongated 2 to 7 times after massive overdosing, explaining why glucose supplementation is needed for long periods in these cases.
Collapse
Affiliation(s)
- Yuka Sato
- Diabetes Center, Aizawa Hospital, Matsumoto, 390-8510, Japan
| | - Yuta Mizuno
- Department of Critical Care and Emergency Medicine, Aizawa Hospital, Matsumoto, 390-8510, Japan
| | - Kazuki Suganuma
- Department of Critical Care and Emergency Medicine, Aizawa Hospital, Matsumoto, 390-8510, Japan
| | - Kosuke Shiroto
- Department of Critical Care and Emergency Medicine, Aizawa Hospital, Matsumoto, 390-8510, Japan
| | - Takeshi Ikeda
- Intensive Care Unit, Aizawa Hospital, Matsumoto, 390-8510, Japan
| | - Koh Yamashita
- Diabetes Center, Aizawa Hospital, Matsumoto, 390-8510, Japan
| | - Toshihide Kimura
- Department of Pharmacology, Oita University Faculty of Medicine, Oita, 879-5533, Japan
| | - Keishi Yamauchi
- Department of Diabetes, Endocrinology and Metabolism, International University of Health and Welfare Shioya Hospital, Yaita, 329-2145, Japan
| | - Toru Aizawa
- Diabetes Center, Aizawa Hospital, Matsumoto, 390-8510, Japan
| |
Collapse
|
31
|
Frank S, Jbaily A, Hinshaw L, Basu R, Basu A, Szeri AJ. Modeling the acute effects of exercise on insulin kinetics in type 1 diabetes. J Pharmacokinet Pharmacodyn 2018; 45:829-845. [PMID: 30392154 DOI: 10.1007/s10928-018-9611-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 10/24/2018] [Indexed: 01/24/2023]
Abstract
Our objective is to develop a physiology-based model of insulin kinetics to understand how exercise alters insulin concentrations in those with type 1 diabetes (T1D). We reveal the relationship between the insulin absorption rate ([Formula: see text]) from subcutaneous tissue, the insulin delivery rate ([Formula: see text]) to skeletal muscle, and two physiological parameters that characterize the tissue: the perfusion rate (Q) and the capillary permeability surface area (PS), both of which increase during exercise because of capillary recruitment. We compare model predictions to experimental observations from two pump-wearing T1D cohorts [resting subjects ([Formula: see text]) and exercising subjects ([Formula: see text])] who were each given a mixed-meal tolerance test and a bolus of insulin. Using independently measured values of Q and PS from literature, the model predicts that during exercise insulin concentration increases by 30% in plasma and by 60% in skeletal muscle. Predictions reasonably agree with experimental observations from the two cohorts, without the need for parameter estimation by curve fitting. The insulin kinetics model suggests that the increase in surface area associated with exercise-induced capillary recruitment significantly increases [Formula: see text] and [Formula: see text], which explains why insulin concentrations in plasma and skeletal muscle increase during exercise, ultimately enhancing insulin-dependent glucose uptake. Preventing hypoglycemia is of paramount importance in determining the proper insulin dose during exercise. The presented model provides mechanistic insight into how exercise affects insulin kinetics, which could be useful in guiding the design of decision support systems and artificial pancreas control algorithms.
Collapse
Affiliation(s)
- Spencer Frank
- Department of Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA.
| | - Abdulrahman Jbaily
- Department of Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA.,Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ling Hinshaw
- Division of Endocrinology, Mayo Clinic, Rochester, MI, USA
| | - Rita Basu
- Division of Endocrinology, Mayo Clinic, Rochester, MI, USA.,Department of Endocrinology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ananda Basu
- Division of Endocrinology, Mayo Clinic, Rochester, MI, USA.,Department of Endocrinology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Andrew J Szeri
- Department of Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA.,Department of Mechanical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Adams GG, Meal A, Morgan PS, Alzahrani QE, Zobel H, Lithgo R, Kok MS, Besong DTM, Jiwani SI, Ballance S, Harding SE, Chayen N, Gillis RB. Characterisation of insulin analogues therapeutically available to patients. PLoS One 2018; 13:e0195010. [PMID: 29596514 PMCID: PMC5875863 DOI: 10.1371/journal.pone.0195010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/14/2018] [Indexed: 11/29/2022] Open
Abstract
The structure and function of clinical dosage insulin and its analogues were assessed. This included ‘native insulins’ (human recombinant, bovine, porcine), ‘fast-acting analogues’ (aspart, glulisine, lispro) and ‘slow-acting analogues’ (glargine, detemir, degludec). Analytical ultracentrifugation, both sedimentation velocity and equilibrium experiments, were employed to yield distributions of both molar mass and sedimentation coefficient of all nine insulins. Size exclusion chromatography, coupled to multi-angle light scattering, was also used to explore the function of these analogues. On ultracentrifugation analysis, the insulins under investigation were found to be in numerous conformational states, however the majority of insulins were present in a primarily hexameric conformation. This was true for all native insulins and two fast-acting analogues. However, glargine was present as a dimer, detemir was a multi-hexameric system, degludec was a dodecamer (di-hexamer) and glulisine was present as a dimer-hexamer-dihexamer system. However, size-exclusion chromatography showed that the two hexameric fast-acting analogues (aspart and lispro) dissociated into monomers and dimers due to the lack of zinc in the mobile phase. This comprehensive study is the first time all nine insulins have been characterised in this way, the first time that insulin detemir have been studied using analytical ultracentrifugation and the first time that insulins aspart and glulisine have been studied using sedimentation equilibrium. The structure and function of these clinically administered insulins is of critical importance and this research adds novel data to an otherwise complex functional physiological protein.
Collapse
Affiliation(s)
- Gary G. Adams
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
- * E-mail: (GGA); (RBG)
| | - Andrew Meal
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Paul S. Morgan
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Qushmua E. Alzahrani
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
- Taif University, Faculty of Science, Taif, Saudi Arabia
| | | | - Ryan Lithgo
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
| | - M. Samil Kok
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- Department of Food Engineering, Abant Izzet Baysal University, Bolu, Turkey
| | - David T. M. Besong
- Functional Nanomaterials Lab, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Shahwar I. Jiwani
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
| | | | - Stephen E. Harding
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
| | - Naomi Chayen
- Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Richard B. Gillis
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
- * E-mail: (GGA); (RBG)
| |
Collapse
|