101
|
A Diphenylalanine Based Pentapeptide with Fibrillating Self-Assembling Properties. Pharmaceutics 2023; 15:pharmaceutics15020371. [PMID: 36839694 PMCID: PMC9966497 DOI: 10.3390/pharmaceutics15020371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Peptides and their related compounds can self-assemble into diverse nanostructures of different shapes and sizes in response to various stimuli such as pH, temperature or ionic strength. Here we report the synthesis and characterization of a lysozyme derived pentapeptide and its ability to build well-defined fibrillar structures. Lysozyme FESNF peptide fragment was synthesized by solid phase peptide synthesis using the Fmoc/t-Bu strategy, purified by analytical high-performance liquid chromatography (HPLC) and its molecular weight was confirmed by matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS). Spectroscopic features of this pentapeptide were investigated by UV-visible spectroscopy and fluorimetry showing the pattern of marginal phenylalanine residues within the peptide sequence. Self-assembling properties were determined using atomic force microscopy (AFM), aggregation index and thioflavin T assay (ThT). FESNF generating fibrillar structures observed by AFM and aggregation propensity were primarily influenced by pH conditions. Moreover, the experimental data were confirmed by molecular dynamics simulation studies. The obtained fibrils will be used next to explore their potential to act as support material for medical and cosmetic application.
Collapse
|
102
|
Chen K, Smith CJ. Best Practices for Submission of NMR Data to Support Higher Order Structure Assessment of Generic Peptide Drugs. AAPS J 2023; 25:17. [PMID: 36670271 DOI: 10.1208/s12248-023-00782-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Affiliation(s)
- Kang Chen
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA.
| | - Cameron J Smith
- Division of Liquid-Based Products I, Office of Lifecycle Drug Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| |
Collapse
|
103
|
Forlano N, Bucci R, Contini A, Venanzi M, Placidi E, Gelmi ML, Lettieri R, Gatto E. Non-Conventional Peptide Self-Assembly into a Conductive Supramolecular Rope. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13020333. [PMID: 36678086 PMCID: PMC9867255 DOI: 10.3390/nano13020333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 05/27/2023]
Abstract
Structures composed of alternating α and β amino acids can give rise to peculiar secondary structural motifs, which could self-assemble into complex structures of controlled geometries. This work describes the self-assembly properties of an α,β-peptide, containing three units of syn H2-(2-F-Phe)-h-PheGly-OH, able to self-organize on surfaces into a fascinating supramolecular rope. This material was characterized by AFM, electronic conduction and fluorescence measurements. Molecular dynamics simulations showed that this hexapeptide can self-assemble into an antiparallel β-sheet layer, stabilized by intermolecular H-bonds, which, in turn, can self-assemble into many side-by-side layers, due to π-π interactions. As a matter of fact, we demonstrated that in this system, the presence of aromatic residues at the intramolecular interface promoted by the alternation of α,β-amino-acids in the primary sequence, endorses the formation of a super-secondary structure where the aromatic groups are close to each other, conferring to the system good electron conduction properties. This work demonstrates the capability and future potential of designing and fabricating distinctive nanostructures and efficient bioelectronic interfaces based on an α,β-peptide, by controlling structure and interaction processes beyond those obtained with α- or β-peptides alone.
Collapse
Affiliation(s)
- Nicola Forlano
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Roma, Italy
| | - Raffaella Bucci
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milan, Italy
| | - Alessandro Contini
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milan, Italy
| | - Mariano Venanzi
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Roma, Italy
| | - Ernesto Placidi
- Department of Physics, Sapienza University of Rome, P.le Aldo Moro 2, 00185 Rome, Italy
| | - Maria Luisa Gelmi
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milan, Italy
| | - Raffaella Lettieri
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Roma, Italy
| | - Emanuela Gatto
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Roma, Italy
| |
Collapse
|
104
|
Frolov AI, Chankeshwara SV, Abdulkarim Z, Ghiandoni GM. pIChemiSt ─ Free Tool for the Calculation of Isoelectric Points of Modified Peptides. J Chem Inf Model 2023; 63:187-196. [PMID: 36573842 PMCID: PMC9832473 DOI: 10.1021/acs.jcim.2c01261] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The isoelectric point (pI) is a fundamental physicochemical property of peptides and proteins. It is widely used to steer design away from low solubility and aggregation and guide peptide separation and purification. Experimental measurements of pI can be replaced by calculations knowing the ionizable groups of peptides and their corresponding pKa values. Different pKa sets are published in the literature for natural amino acids, however, they are insufficient to describe synthetically modified peptides, complex peptides of natural origin, and peptides conjugated with structures of other modalities. Noncanonical modifications (nCAAs) are ignored in the conventional sequence-based pI calculations, therefore producing large errors in their pI predictions. In this work, we describe a pI calculation method that uses the chemical structure as an input, automatically identifies ionizable groups of nCAAs and other fragments, and performs pKa predictions for them. The method is validated on a curated set of experimental measures on 29 modified and 119093 natural peptides, providing an improvement of R2 from 0.74 to 0.95 and 0.96 against the conventional sequence-based approach for modified peptides for the two studied pKa prediction tools, ACDlabs and pKaMatcher, correspondingly. The method is available in the form of an open source Python library at https://github.com/AstraZeneca/peptide-tools, which can be integrated into other proprietary and free software packages. We anticipate that the pI calculation tool may facilitate optimization and purification activities across various application domains of peptides, including the development of biopharmaceuticals.
Collapse
Affiliation(s)
- Andrey I. Frolov
- Medicinal
Chemistry, Research and Early Development, Cardiovascular, Renal and
Metabolism (CVRM), BioPharmaceuticals R&D,
AstraZeneca, Gothenburg, Sweden,
| | - Sunay V. Chankeshwara
- Medicinal
Chemistry, Research and Early Development, Cardiovascular, Renal and
Metabolism (CVRM), BioPharmaceuticals R&D,
AstraZeneca, Gothenburg, Sweden
| | - Zeyed Abdulkarim
- Early
Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
105
|
Klich JH, Kasse CM, Mann JL, Huang Y, d’Aquino AI, Grosskopf AK, Baillet J, Fuller GG, Appel EA. Stable High-Concentration Monoclonal Antibody Formulations Enabled by an Amphiphilic Copolymer Excipient. ADVANCED THERAPEUTICS 2023; 6:2200102. [PMID: 36684707 PMCID: PMC9854243 DOI: 10.1002/adtp.202200102] [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] [Indexed: 01/26/2023]
Abstract
Monoclonal antibodies are a staple in modern pharmacotherapy. Unfortunately, these biopharmaceuticals are limited by their tendency to aggregate in formulation, resulting in poor stability and often requiring low concentration drug formulations. Moreover, existing excipients designed to stabilize these formulations are often limited by their toxicity and tendency to form particles such as micelles. Here, we demonstrate the ability of a simple "drop-in", amphiphilic copolymer excipient to enhance the stability of high concentration formulations of clinically-relevant monoclonal antibodies without altering their pharmacokinetics or injectability. Through interfacial rheology and surface tension measurements, we demonstrate that the copolymer excipient competitively adsorbs to formulation interfaces. Further, through determination of monomeric composition and retained bioactivity through stressed aging, we show that this excipient confers a significant stability benefit to high concentration antibody formulations. Finally, we demonstrate that the excipient behaves as an inactive ingredient, having no significant impact on the pharmacokinetic profile of a clinically relevant antibody in mice. This amphiphilic copolymer excipient demonstrates promise as a simple formulation additive to create stable, high concentration antibody formulations, thereby enabling improved treatment options such as a route-of-administration switch from low concentration intravenous (IV) to high concentration subcutaneous (SC) delivery while reducing dependence on the cold chain.
Collapse
Affiliation(s)
| | | | - Joseph L. Mann
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Yaoqi Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Andrea I. d’Aquino
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Abigail K. Grosskopf
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Julie Baillet
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Gerald G. Fuller
- Department of Chemical Engineering, 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 School of Medicine, Stanford, CA 94305, USA; ChEM-H Institute, Stanford University, Stanford, CA 94305, USA; Woods Institute for the Environment, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
106
|
Kurtzhals P, Østergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov 2023; 22:59-80. [PMID: 36002588 DOI: 10.1038/s41573-022-00529-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 01/28/2023]
Abstract
Peptides and proteins are widely used to treat a range of medical conditions; however, they often have to be injected and their effects are short-lived. These shortcomings of the native structure can be addressed by molecular engineering, but this is a complex undertaking. A molecular engineering technology initially applied to insulin - and which has now been successfully applied to several biopharmaceuticals - entails the derivatization of peptides and proteins with fatty acids. Various protraction mechanisms are enabled by the specific characteristics and positions of the attached fatty acid. Furthermore, the technology can ensure a long half-life following oral administration of peptide drugs, can alter the distribution of peptides and may hold potential for tissue targeting. Due to the inherent safety and well-defined chemical nature of the fatty acids, this technology provides a versatile approach to peptide and protein drug discovery.
Collapse
|
107
|
Vakili S, Chamani J, Amiri-Tehranizadeh Z, Hosseinzadeh H, Mosaffa F, Khatami SM, Khameneh B, Saberi MR. Amino acid-mPEGs: Promising excipients to stabilize human growth hormone against aggregation. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:635-644. [PMID: 37275756 PMCID: PMC10237159 DOI: 10.22038/ijbms.2023.67557.14804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 03/07/2023] [Indexed: 06/07/2023]
Abstract
Objectives Today, the non-covalent PEGylation methods of protein pharmaceuticals attract more attention and possess several advantages over the covalent approach. In the present study, Amino Acid-mPEGs (aa-mPEGs) were synthesized, and the human Growth Hormone (hGH) stability profile was assessed in their presence and absence. Materials and Methods aa-mPEGs were synthesized with different amino acids (Trp, Glu, Arg, Cys, and Leu) and molecular weights of polymers (2 and 5 KDa). The aa-mPEGs were analyzed with different methods. The physical and structural stabilities of hGH were analyzed by SEC and CD spectroscopy methods. Physical stability was assayed at different temperatures within certain intervals. Molecular dynamics (MD) simulation was used to realize the possible mode of interaction between protein and aa-mPEGs. The cell-based method was used to evaluate the cytotoxicity. Results HNMR and FTIR spectroscopy indicated that aa-mPEGs were successfully synthesized. hGH as a control group is known to be stable at 4 °C; a pronounced change in monomer degradation is observed when stored at 25 °C and 37 °C. hGH:Glu-mPEG 2 kDa with a molar ratio of 1:1 to the protein solution can significantly increase the physical stability. The CD spectroscopy method showed that the secondary structure of the protein was preserved during storage. aa-mPEGs did not show any cytotoxicity activities. The results of MD simulations were in line with experimental results. Conclusion This paper showed that aa-mPEGs are potent excipients in decreasing the aggregation of hGH. Glu-mPEG exhibited the best-stabilizing properties in a harsh environment among other aa-mPEGs.
Collapse
Affiliation(s)
- Somayeh Vakili
- Department of Medical Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - JamshidKhan Chamani
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Zeinab Amiri-Tehranizadeh
- Department of Medical Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Mola Khatami
- Department of Chemistry, Faculty of Samen Hojaj, Mashhad Branch, Technical and Vocational University (TVU), Tehran, Iran
| | - Bahman Khameneh
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Saberi
- Department of Medical Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
108
|
Khan AN, Khan RH. Protein misfolding and related human diseases: A comprehensive review of toxicity, proteins involved, and current therapeutic strategies. Int J Biol Macromol 2022; 223:143-160. [PMID: 36356861 DOI: 10.1016/j.ijbiomac.2022.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Most of the cell's chemical reactions and structural components are facilitated by proteins. But proteins are highly dynamic molecules, where numerous modifications or changes in the cellular environment can affect their native conformational fold leading to protein aggregation. Various stress conditions, such as oxidative stress, mutations and metal toxicity may cause protein misfolding and aggregation by shifting the conformational equilibrium towards more aggregation-prone states. Most of the protein misfolding diseases (PMDs) involve aggregation of protein. We have discussed such proteins like Aβ peptide, α-synuclein, amylin and lysozyme involved in Alzheimer's, Parkinson's, type II diabetes and non-neuropathic systemic amyloidosis respectively. Till date, all advances in PMDs therapeutics help symptomatically but do not prevent the root cause of the disease, i.e., the aggregation of protein involved in the diseases. Current efforts focused on developing therapies for PMDs have employed diverse strategies; repositioning pre-existing drugs as it saves time and money; natural compounds that are touted as potential drug candidates have an advantage of being taken in diet normally and will induce lesser side effects. This review also covers recently developed therapeutic strategies like antisense drugs and disaggregases which has yielded therapeutic agents that have transitioned from preclinical studies into human clinical trials.
Collapse
Affiliation(s)
- Asra Nasir Khan
- Interdisciplinary Biotechnology Unit, AMU, Aligarh 202002, India
| | | |
Collapse
|
109
|
Studying Peptide-Metal Ion Complex Structures by Solution-State NMR. Int J Mol Sci 2022; 23:ijms232415957. [PMID: 36555599 PMCID: PMC9782655 DOI: 10.3390/ijms232415957] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Metal chelation can provide structural stability and form reactive centers in metalloproteins. Approximately one third of known protein structures are metalloproteins, and metal binding, or the lack thereof, is often implicated in disease, making it necessary to be able to study these systems in detail. Peptide-metal complexes are both present in nature and can provide a means to focus on the binding region of a protein and control experimental variables to a high degree. Structural studies of peptide complexes with metal ions by nuclear magnetic resonance (NMR) were surveyed for all the essential metal complexes and many non-essential metal complexes. The various methods used to study each metal ion are presented together with examples of recent research. Many of these metal systems have been individually reviewed and this current overview of NMR studies of metallopeptide complexes aims to provide a basis for inspiration from structural studies and methodology applied in the field.
Collapse
|
110
|
Hemmati S, Rasekhi Kazerooni H. Polypharmacological Cell-Penetrating Peptides from Venomous Marine Animals Based on Immunomodulating, Antimicrobial, and Anticancer Properties. Mar Drugs 2022; 20:md20120763. [PMID: 36547910 PMCID: PMC9787916 DOI: 10.3390/md20120763] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/09/2022] Open
Abstract
Complex pathological diseases, such as cancer, infection, and Alzheimer's, need to be targeted by multipronged curative. Various omics technologies, with a high rate of data generation, demand artificial intelligence to translate these data into druggable targets. In this study, 82 marine venomous animal species were retrieved, and 3505 cryptic cell-penetrating peptides (CPPs) were identified in their toxins. A total of 279 safe peptides were further analyzed for antimicrobial, anticancer, and immunomodulatory characteristics. Protease-resistant CPPs with endosomal-escape ability in Hydrophis hardwickii, nuclear-localizing peptides in Scorpaena plumieri, and mitochondrial-targeting peptides from Synanceia horrida were suitable for compartmental drug delivery. A broad-spectrum S. horrida-derived antimicrobial peptide with a high binding-affinity to bacterial membranes was an antigen-presenting cell (APC) stimulator that primes cytokine release and naïve T-cell maturation simultaneously. While antibiofilm and wound-healing peptides were detected in Synanceia verrucosa, APC epitopes as universal adjuvants for antiviral vaccination were in Pterois volitans and Conus monile. Conus pennaceus-derived anticancer peptides showed antiangiogenic and IL-2-inducing properties with moderate BBB-permeation and were defined to be a tumor-homing peptide (THP) with the ability to inhibit programmed death ligand-1 (PDL-1). Isoforms of RGD-containing peptides with innate antiangiogenic characteristics were in Conus tessulatus for tumor targeting. Inhibitors of neuropilin-1 in C. pennaceus are proposed for imaging probes or therapeutic delivery. A Conus betulinus cryptic peptide, with BBB-permeation, mitochondrial-targeting, and antioxidant capacity, was a stimulator of anti-inflammatory cytokines and non-inducer of proinflammation proposed for Alzheimer's. Conclusively, we have considered the dynamic interaction of cells, their microenvironment, and proportional-orchestrating-host- immune pathways by multi-target-directed CPPs resembling single-molecule polypharmacology. This strategy might fill the therapeutic gap in complex resistant disorders and increase the candidates' clinical-translation chance.
Collapse
Affiliation(s)
- Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Correspondence: ; Tel.: +98-7132-424-128
| | | |
Collapse
|
111
|
Lawson KE, Dekle JK, Adamczyk AJ. Towards pharmaceutical protein stabilization: DFT and statistical learning studies on non-enzymatic peptide hydrolysis degradation mechanisms. COMPUT THEOR CHEM 2022. [DOI: 10.1016/j.comptc.2022.113938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
112
|
Bana AAK, Mehta P, Ramnani KAK. Physical Instabilities of Therapeutic Monoclonal Antibodies: A Critical Review. Curr Drug Discov Technol 2022; 19:e240622206367. [PMID: 35748546 DOI: 10.2174/1570163819666220624092622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 01/27/2023]
Abstract
The proteinaceous nature of monoclonal antibodies (mAbs) makes them highly sensitive to various physical and chemical conditions, thus leading to instabilities that are classified as physical and chemical instabilities. In this review, we are discussing in detail the physical instability of mAbs because a large number of articles previously published solely focus on the chemical aspect of the instability with little coverage on the physical side. The physical instabilities of mAbs are classified into denaturation and aggregation (precipitation, visible and subvisible particles). The mechanism involved in their formation is discussed in the article, along with the pathways correlating the denaturation of mAb or the formation of aggregates to immunogenicity. Further equations like Gibbs-Helmholtz involved in detecting and quantifying denaturation are discussed, along with various factors causing the denaturation. Moreover, questions related to aggregation like the types of aggregates and the pathway involved in their formation are answered in this article. Factors influencing the physical stability of the mAbs by causing denaturation or formation of aggregates involving the structure of the protein, concentration of mAbs, pH of the protein and the formulations, excipients involved in the formulations, salts added to the formulations, storage temperature, light and UV radiation exposure and processing factors are mentioned in this article. Finally, the analytical approaches used for detecting and quantifying the physical instability of mAbs at all levels of structural conformation like far and near UV, infrared spectroscopy, capillary electrophoresis, LC-MS, microflow imagining, circular dichroism and peptide mapping are discussed.
Collapse
Affiliation(s)
- Arpit Arun K Bana
- Department of Pharmaceutical Analysis, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | - Priti Mehta
- Department of Pharmaceutical Analysis, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | | |
Collapse
|
113
|
El Hauadi K, Resina L, Zanuy D, Esteves T, Ferreira FC, Pérez-Madrigal MM, Alemán C. Dendritic Self-assembled Structures from Therapeutic Charged Pentapeptides. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:12905-12914. [PMID: 36229043 PMCID: PMC9988208 DOI: 10.1021/acs.langmuir.2c02010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/04/2022] [Indexed: 06/16/2023]
Abstract
CRENKA [Cys-Arg-(NMe)Glu-Lys-Ala, where (NMe)Glu refers to N-methyl-Glu], an anti-cancer pentapeptide that induces prostate tumor necrosis and significant reduction in tumor growth, was engineered to increase the resistance to endogenous proteases of its parent peptide, CREKA (Cys-Arg-Glu-Lys-Ala). Considering their high tendency to aggregate, the self-assembly of CRENKA and CREKA into well-defined and ordered structures has been examined as a function of peptide concentration and pH. Spectroscopic studies and atomistic molecular dynamics simulations reveal significant differences between the secondary structures of CREKA and CRENKA. Thus, the restrictions imposed by the (NMe)Glu residue reduce the conformational variability of CRENKA with respect to CREKA, which significantly affects the formation of well-defined and ordered self-assembly morphologies. Aggregates with poorly defined morphology are obtained from solutions with low and moderate CREKA concentrations at pH 4, whereas well-defined dendritic microstructures with fractal geometry are obtained from CRENKA solutions with similar peptide concentrations at pH 4 and 7. The formation of dendritic structures is proposed to follow a two-step mechanism: (1) pseudo-spherical particles are pre-nucleated through a diffusion-limited aggregation process, pre-defining the dendritic geometry, and (2) such pre-nucleated structures coalesce by incorporating conformationally restrained CRENKA molecules from the solution to their surfaces, forming a continuous dendritic structure. Instead, no regular assembly is obtained from solutions with high peptide concentrations, as their dynamics is dominated by strong repulsive peptide-peptide electrostatic interactions, and from solutions at pH 10, in which the total peptide charge is zero. Overall, results demonstrate that dendritic structures are only obtained when the molecular charge of CRENKA, which is controlled through the pH, favors kinetics over thermodynamics during the self-assembly process.
Collapse
Affiliation(s)
- Karima El Hauadi
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
| | - Leonor Resina
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
- Department
of Bioengineering, iBB − Institute for Bioengineering and Biosciences,
Instituto Superior Técnico, Universidade
de Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB—Institute for Health and Bioeconomy at Instituto
Superior Técnico, Universidade de
Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
| | - David Zanuy
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
| | - Teresa Esteves
- Department
of Bioengineering, iBB − Institute for Bioengineering and Biosciences,
Instituto Superior Técnico, Universidade
de Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB—Institute for Health and Bioeconomy at Instituto
Superior Técnico, Universidade de
Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
| | - Frederico Castelo Ferreira
- Department
of Bioengineering, iBB − Institute for Bioengineering and Biosciences,
Instituto Superior Técnico, Universidade
de Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB—Institute for Health and Bioeconomy at Instituto
Superior Técnico, Universidade de
Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
| | - Maria M. Pérez-Madrigal
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
| | - Carlos Alemán
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain
| |
Collapse
|
114
|
Kommineni N, Butreddy A, Sainaga Jyothi VG, Angsantikul P. Freeze-drying for the preservation of immunoengineering products. iScience 2022; 25:105127. [PMID: 36267916 PMCID: PMC9576584 DOI: 10.1016/j.isci.2022.105127] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Immunoengineering technologies harness the power of immune system modulators such as monoclonal antibodies, cytokines, and vaccines to treat myriad diseases. Immunoengineering innovations have showed great promise in various practices including oncology, infectious disease, autoimmune diseases, and transplantation. Despite the countless successes, the majority of immunoengineering products contain active moieties that are prone to instability. The current review aims to feature freeze-drying as a robust and scalable solution to the inherent stability challenges in immunoengineering products by preventing the active moiety from degradation. Furthermore, this review describes the stability issues related to immunoengineering products and the utility of the lyophilization process to preserve the integrity and efficacy of immunoengineering tools ranging from biologics to nanoparticle-based vaccines. The concept of the freeze-drying process is described highlighting the quality by design (QbD) for robust process optimization. Case studies of lyophilized immunoengineering technologies and relevant clinical studies using immunoengineering products are discussed.
Collapse
Affiliation(s)
| | - Arun Butreddy
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
| | - Vaskuri G.S. Sainaga Jyothi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037, India
| | | |
Collapse
|
115
|
Antiangiogenic potential of small polypeptide sequences: In vivo assays, cytotoxicity, synthetic approaches and influence of C-terminal carboxyamidation. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
116
|
Schifano NP, Caputo GA. Investigation of the Role of Hydrophobic Amino Acids on the Structure-Activity Relationship in the Antimicrobial Venom Peptide Ponericin L1. J Membr Biol 2022; 255:537-551. [PMID: 34792624 PMCID: PMC9114170 DOI: 10.1007/s00232-021-00204-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022]
Abstract
Venom mixtures from insects, reptiles, and mollusks have long been a source of bioactive peptides which often have alternative uses as therapeutics. While these molecules act in numerous capacities, there have been many venom components that act on the target cells through membrane disruptive mechanisms. These peptides have long been of interest as potential antimicrobial peptide platforms, but the inherent cytotoxicity of venom peptides often results in poor therapeutic potential. Despite this, efforts are ongoing to identify and characterize venom peptide which exhibit high antimicrobial activity with low cytotoxicity and modify these to further enhance the efficacy while reducing toxicity. One example is ponericin L1 from Neoponera goeldii which has been demonstrated to have good antimicrobial activity and low in vitro cytotoxicity. The L1 sequence was modified by uniformly replacing the native hydrophobic residues with either Leu, Ile, Phe, Ala, or Val. Spectroscopic and microbiological approaches were employed to investigate how the amino acid sequence changes impacted membrane interaction, secondary structure, and antimicrobial efficacy. The L1 derivatives showed varying degrees of bilayer interaction, in some cases driven by bilayer composition. Several of the variants exhibited enhanced antimicrobial activity compared to the parent strain, while others lost all activity. Interestingly, the variant containing Val lost all antimicrobial activity and ability to interact with bilayers. Taken together the results indicate that peptide secondary structure, amino acid composition, and hydrophobicity all play a role in peptide activity, although this is a delicate balance that can result in non-specific binding or complete loss of activity if specific amino acids are incorporated.
Collapse
Affiliation(s)
- Nicholas P Schifano
- Department of Chemistry & Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, NJ, 08028, USA
| | - Gregory A Caputo
- Department of Chemistry & Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, NJ, 08028, USA.
- Department of Molecular & Cellular Biosciences, Rowan University, 201 Mullica Hill Road, Glassboro, NJ, 08028, USA.
| |
Collapse
|
117
|
Baidya A, Haghniaz R, Tom G, Edalati M, Kaneko N, Alizadeh P, Tavafoghi M, Khademhosseini A, Sheikhi A. A Cohesive Shear-Thinning Biomaterial for Catheter-Based Minimally Invasive Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42852-42863. [PMID: 36121372 DOI: 10.1021/acsami.2c08799] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Shear-thinning hydrogels are suitable biomaterials for catheter-based minimally invasive therapies; however, the tradeoff between injectability and mechanical integrity has limited their applications, particularly at high external shear stress such as that during endovascular procedures. Extensive molecular crosslinking often results in stiff, hard-to-inject hydrogels that may block catheters, whereas weak crosslinking renders hydrogels mechanically weak and susceptible to shear-induced fragmentation. Thus, controlling molecular interactions is necessary to improve the cohesion of catheter-deployable hydrogels. To address this material design challenge, we have developed an easily injectable, nonhemolytic, and noncytotoxic shear-thinning hydrogel with significantly enhanced cohesion via controlling noncovalent interactions. We show that enhancing the electrostatic interactions between weakly bound biopolymers (gelatin) and nanoparticles (silicate nanoplatelets) using a highly charged polycation at an optimum concentration increases cohesion without compromising injectability, whereas introducing excessive charge to the system leads to phase separation and loss of function. The cohesive biomaterial is successfully injected with a neuroendovascular catheter and retained without fragmentation in patient-derived three-dimensionally printed cerebral aneurysm models under a physiologically relevant pulsatile fluid flow, which would otherwise be impossible using the noncohesive hydrogel counterpart. This work sheds light on how charge-driven molecular and colloidal interactions in shear-thinning physical hydrogels improve cohesion, enabling complex minimally invasive procedures under flow, which may open new opportunities for developing the next generation of injectable biomaterials.
Collapse
Affiliation(s)
- Avijit Baidya
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Reihaneh Haghniaz
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California 90024, United States
| | - Gregory Tom
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
| | - Masoud Edalati
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Naoki Kaneko
- Division of Interventional Neuroradiology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Parvin Alizadeh
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Maryam Tavafoghi
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
| | - Ali Khademhosseini
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California 90024, United States
| | - Amir Sheikhi
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
118
|
McMackin P, Adam J, Griffin S, Hirsa A. Amyloidogenesis via interfacial shear in a containerless biochemical reactor aboard the International Space Station. NPJ Microgravity 2022; 8:41. [PMID: 36127358 PMCID: PMC9489778 DOI: 10.1038/s41526-022-00227-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022] Open
Abstract
Fluid interfaces significantly influence the dynamics of protein solutions, effects that can be isolated by performing experiments in microgravity, greatly reducing the amount of solid boundaries present, allowing air-liquid interfaces to become dominant. This investigation examined the effects of protein concentration on interfacial shear-induced fibrillization of insulin in microgravity within a containerless biochemical reactor, the ring-sheared drop (RSD), aboard the international space station (ISS). Human insulin was used as a model amyloidogenic protein for studying protein kinetics with applications to in situ pharmaceutical production, tissue engineering, and diseases such as Alzheimer’s, Parkinson’s, infectious prions, and type 2 diabetes. Experiments investigated three main stages of amyloidogenesis: nucleation studied by seeding native solutions with fibril aggregates, fibrillization quantified using intrinsic fibrillization rate after fitting measured solution intensity to a sigmoidal function, and gelation observed by detection of solidification fronts. Results demonstrated that in surface-dominated amyloidogenic protein solutions: seeding with fibrils induces fibrillization of native protein, intrinsic fibrillization rate is independent of concentration, and that there is a minimum fibril concentration for gelation with gelation rate and rapidity of onset increasing monotonically with increasing protein concentration. These findings matched well with results of previous studies within ground-based analogs.
Collapse
Affiliation(s)
- Patrick McMackin
- Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA
| | - Joe Adam
- Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA.,Department of Biological Sciences, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA.,Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA
| | - Shannon Griffin
- Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA.,Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA
| | - Amir Hirsa
- Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA. .,Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA.
| |
Collapse
|
119
|
Mun SJ, Cho E, Kim JS, Yang CS. Pathogen-derived peptides in drug targeting and its therapeutic approach. J Control Release 2022; 350:716-733. [PMID: 36030988 DOI: 10.1016/j.jconrel.2022.08.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 02/06/2023]
Abstract
Peptides, short stretches of amino acids or small proteins that occupy a strategic position between proteins and amino acids, are readily accessible by chemical and biological methods. With ideal properties for forming high-affinity and specific interactions with host target proteins, they have established an important niche in the drug development spectrum complementing small molecule and biological therapeutics. Among the most successful biomedicines in use today, peptide-based drugs show great promise. This, coupled with recent advances in synthetic and nanochemical biology, has led to the creation of tailor-made peptide therapeutics for improved biocompatibility. This review presents an overview of the latest research on pathogen-derived, host-cell-interacting peptides. It also highlights strategies for using peptide-based therapeutics that address cellular transport challenges through the introduction of nanoparticles that serve as platforms to facilitate the delivery of peptide biologics and therapeutics for treating various inflammatory diseases. Finally, this paper describes future perspectives, specific pathogen-based peptides that can enhance specificity, efficiency, and capacity in functional peptide-based therapeutics, which are in the spotlight as new treatment alternatives for various diseases, and also presents verified sequences and targets that can increase chemical and pharmacological value.
Collapse
Affiliation(s)
- Seok-Jun Mun
- Department of Bionano Technology, Hanyang University, Seoul 04673, Republic of Korea; Center for Bionano Intelligence Education and Research, Ansan 15588, Republic of Korea
| | - Euni Cho
- Department of Bionano Technology, Hanyang University, Seoul 04673, Republic of Korea; Center for Bionano Intelligence Education and Research, Ansan 15588, Republic of Korea
| | - Jae-Sung Kim
- Department of Bionano Technology, Hanyang University, Seoul 04673, Republic of Korea; Institute of Natural Science & Technology, Hanyang University, Ansan 15588, Republic of Korea
| | - Chul-Su Yang
- Center for Bionano Intelligence Education and Research, Ansan 15588, Republic of Korea; Department of Molecular and Life Science, Hanyang University, Ansan 15588, Republic of Korea.
| |
Collapse
|
120
|
Mielecki M, Ziemniak M, Ozga M, Borowski R, Antosik J, Kaczyńska A, Pająk B. Structure-Activity Relationship of the Dimeric and Oligomeric Forms of a Cytotoxic Biotherapeutic Based on Diphtheria Toxin. Biomolecules 2022; 12:biom12081111. [PMID: 36009005 PMCID: PMC9406121 DOI: 10.3390/biom12081111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Protein aggregation is a well-recognized problem in industrial preparation, including biotherapeutics. These low-energy states constantly compete with a native-like conformation, which is more pronounced in the case of macromolecules of low stability in the solution. A better understanding of the structure and function of such aggregates is generally required for the more rational development of therapeutic proteins, including single-chain fusion cytotoxins to target specific receptors on cancer cells. Here, we identified and purified such particles as side products of the renaturation process of the single-chain fusion cytotoxin, composed of two diphtheria toxin (DT) domains and interleukin 13 (IL-13), and applied various experimental techniques to comprehensively understand their molecular architecture and function. Importantly, we distinguished soluble purified dimeric and fractionated oligomeric particles from aggregates. The oligomers are polydisperse and multimodal, with a distribution favoring lower and even stoichiometries, suggesting they are composed of dimeric building units. Importantly, all these oligomeric particles and the monomer are cystine-dependent as their innate disulfide bonds have structural and functional roles. Their reduction triggers aggregation. Presumably the dimer and lower oligomers represent the metastable state, retaining the native disulfide bond. Although significantly reduced in contrast to the monomer, they preserve some fraction of bioactivity, manifested by their IL-13RA2 receptor affinity and selective cytotoxic potency towards the U-251 glioblastoma cell line. These molecular assemblies probably preserve structural integrity and native-like fold, at least to some extent. As our study demonstrated, the dimeric and oligomeric cytotoxin may be an exciting model protein, introducing a new understanding of its monomeric counterpart’s molecular characteristics.
Collapse
|
121
|
Li M, Falk BT, Lu X, Schroder R, Mccoy M, Xu W, Yin DH, Gindy ME, D'Addio SM, Su Y. Molecular Mechanism of Antimicrobial Excipient-Induced Aggregation in Parenteral Formulations of Peptide Therapeutics. Mol Pharm 2022; 19:3267-3278. [PMID: 35917158 DOI: 10.1021/acs.molpharmaceut.2c00449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antimicrobial preservatives are used as functional excipients in multidose formulations of biological therapeutics to destroy or inhibit the growth of microbial contaminants, which may be introduced by repeatedly administering doses. Antimicrobial agents can also induce the biophysical instability of proteins and peptides, which presents a challenge in optimizing the drug product formulation. Elucidating the structural basis for aggregation aids in understanding the underlying mechanism and can offer valuable knowledge and rationale for designing drug substances and drug products; however, this remains largely unexplored due to the lack of high-resolution characterization. In this work, we utilize solution nuclear magnetic resonance (NMR) as an advanced biophysical tool to study an acylated 31-residue peptide, acyl-peptide A, and its interaction with commonly used antimicrobial agents, benzyl alcohol and m-cresol. Our results suggest that acyl-peptide A forms soluble octamers in the aqueous solution, which tumble slowly due to an increased molecular weight as measured by diffusion ordered spectroscopy and 1H relaxation measurement. The addition of benzyl alcohol does not induce aggregation of acyl-peptide A and has no chemical shift perturbation in 1H-1H NOESY spectra, suggesting no detectable interaction with the peptide. In contrast, the addition of 1% (w/v) m-cresol results in insoluble aggregates composed of 25% (w/w) peptides after a 24-hour incubation at room temperature as quantified by 1H NMR. Interestingly, 1H-13C heteronuclear single-quantum coherence and 1H-1H total correlation experiment spectroscopy have identified m-cresol and peptide interactions at specific residues, including Met, Lys, Glu, and Gln, suggesting that there may be a combination of hydrophobic, hydrogen bonding, and electrostatic interactions with m-cresol driving this phenomenon. These site-specific interactions have promoted the formation of higher-order oligomerization such as dimers and trimers of octamers, eventually resulting in insoluble aggregates. Our study has elucidated a structural basis of m-cresol-induced self-association that can inform the optimized design of drug substances and products. Moreover, it has demonstrated solution NMR as a high-resolution tool to investigate the structure and dynamics of biological drug products and provide an understanding of excipient-induced peptide and protein aggregation.
Collapse
Affiliation(s)
- Mingyue Li
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Bradley T Falk
- Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Xingyu Lu
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ryan Schroder
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Mark Mccoy
- Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Wei Xu
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel H Yin
- Pharmaceutical Sciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Marian E Gindy
- Small Molecule Science and Technology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Suzanne M D'Addio
- Pharmaceutical Sciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yongchao Su
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
122
|
Vince MJK, Holub JM. Synthesis of Scyllatoxin-Based BH3 Domain Mimetics with Diverse Patterns of Native Disulfide Bonds. Curr Protoc 2022; 2:e526. [PMID: 35994574 DOI: 10.1002/cpz1.526] [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: 06/15/2023]
Abstract
This article outlines the design and development of scyllatoxin (ScTx)-based BH3 domain mimetics with diverse patterns of native disulfide bonds. More specifically, this method summarizes the total chemical synthesis of ScTx-based peptides that contain zero, one, two, or three disulfide linkages, including techniques to generate variants with any combination of native disulfides. Each peptide reported herein is generated on solid-phase support using microwave-assisted coupling procedures, and all reaction parameters related to the peptide synthesis are described in detail. The various disulfide patterns of the ScTx-based constructs are established during peptide synthesis and are ultimately verified by mass analysis of trypsin-digested fragments. The BH3 domain mimetics developed herein were generated by transposing residues from the helical BH3 domain of the pro-apoptotic BCL2 protein Bax to the α-helix of wild-type ScTx. Interestingly, we found that the relative binding affinities of ScTx-Bax peptides for the anti-apoptotic BCL2 protein Bcl-2 (proper) were heavily influenced by the number and position of disulfide linkages within the ScTx-Bax sequence. As a consequence, we were able to utilize ScTx-Bax BH3 domain mimetics with varied patterns of disulfide bonds to survey how structural rigidity within the helical Bax BH3 domain affects binding to promiscuous anti-apoptotic BCL2 proteins. More broadly, the ability to generate ScTx-based molecules that contain any combination of native disulfide bonds expands the utility of such constructs as tools to study the molecular nature of protein-protein interactions. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Synthesis and characterization of ScTx-based Bax BH3 domain mimetics Basic Protocol 2: Oxidation of ScTx-Bax BH3 domain mimetics containing one, two, or three disulfide linkages Support Protocol: Mapping of disulfide linkages in oxidized ScTx-Bax BH3 domain mimetics.
Collapse
Affiliation(s)
- Matthew J K Vince
- Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio
- Institut für Bioanalytische Chemie, Fakultät für Chemie und Mineralogie, Universität Leipzig, Leipzig, Germany
- Biotechnologisch-Biomedizinisches Zentrum, Universität Leipzig, Leipzig, Germany
| | - Justin M Holub
- Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Molecular and Cellular Biology Program, Ohio University, Athens, Ohio
| |
Collapse
|
123
|
Oosting LT, Franke K, Martin MV, Kloosterman WP, Jamieson JA, Glenn LA, de Jager MW, van Zanten J, Allersma DP, Gareb B. Development of a Personalized Tumor Neoantigen Based Vaccine Formulation (FRAME-001) for Use in a Phase II Trial for the Treatment of Advanced Non-Small Cell Lung Cancer. Pharmaceutics 2022; 14:pharmaceutics14071515. [PMID: 35890409 PMCID: PMC9322189 DOI: 10.3390/pharmaceutics14071515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 01/27/2023] Open
Abstract
Stage III–IV non-small cell lung cancer (NSCLC) is a devastating disease characterized by a poor prognosis. NSCLC tumors carry genetic mutations, which can lead to the expression of altered protein sequences. Peptides originating from mutated proteins and bound to MHC molecules on the tumor cell surface are referred to as neoantigens, as they are tumor-specific and not expressed in normal cells. Due to their tumor specificity, neoantigens have a strong potential to induce an anti-tumor immune response and have been investigated for development of personalized therapeutic cancer vaccines. The current study describes the development of a clinical grade neoantigen vaccine formulation (FRAME-001) intended as immunotherapy in advanced NSCLC in combination with the immune checkpoint inhibitor pembrolizumab. The detection of aberrant tumor-specific transcripts as well as an algorithm to select immunogenic neoantigen peptides are described. Subsequently, selected neoantigen peptides were synthesized with a high throughput synthesis platform and aseptically formulated under good manufacturing practice (GMP) conditions into four aqueous peptides mixtures that each contained six neoantigen peptides. A validated stability-indicating analytical method was developed in which we considered the personalized nature of the formulation. An extensive stability study performed either at −25 °C or −80 °C showed that the formulation was stable for up to 32 weeks. The formulation was mixed with the vaccine adjuvant Montanide ISA 51 VG, which yielded the final vaccine emulsion. The stability of the vaccine emulsion was demonstrated using microscopic examination, differential light scattering, and the water-drop test. The presented data show that FRAME-001 is a feasible personalized vaccine formulation for the treatment of stage III–IV NSCLC. The presented data may give guidance in the development of novel personalized therapeutic vaccines since this formulation strategy could be used for any cancer indication.
Collapse
Affiliation(s)
- Linette T. Oosting
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.T.O.); (J.v.Z.); (D.P.A.)
| | - Katka Franke
- CureVac Netherlands B.V., Matrix Building VII, Science Park 106, 1098 XG Amsterdam, The Netherlands; (K.F.); (M.V.M.); (W.P.K.)
| | - Michael V. Martin
- CureVac Netherlands B.V., Matrix Building VII, Science Park 106, 1098 XG Amsterdam, The Netherlands; (K.F.); (M.V.M.); (W.P.K.)
| | - Wigard P. Kloosterman
- CureVac Netherlands B.V., Matrix Building VII, Science Park 106, 1098 XG Amsterdam, The Netherlands; (K.F.); (M.V.M.); (W.P.K.)
| | - Jennifer A. Jamieson
- Almac Sciences Scotland Ltd., The Fleming Building, Edinburgh Technopole, Milton Bridge, Penicuik EH26 0BE, UK; (J.A.J.); (L.A.G.)
| | - Laura A. Glenn
- Almac Sciences Scotland Ltd., The Fleming Building, Edinburgh Technopole, Milton Bridge, Penicuik EH26 0BE, UK; (J.A.J.); (L.A.G.)
| | | | - Jacoba van Zanten
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.T.O.); (J.v.Z.); (D.P.A.)
| | - Derk P. Allersma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.T.O.); (J.v.Z.); (D.P.A.)
| | - Bahez Gareb
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.T.O.); (J.v.Z.); (D.P.A.)
- Correspondence:
| |
Collapse
|
124
|
Stable Dried Catalase Particles Prepared by Electrospraying. NANOMATERIALS 2022; 12:nano12142484. [PMID: 35889708 PMCID: PMC9322511 DOI: 10.3390/nano12142484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022]
Abstract
Therapeutic proteins and peptides are clinically important, offering potency while reducing the potential for off-target effects. Research interest in developing therapeutic polypeptides has grown significantly during the last four decades. However, despite the growing research effort, maintaining the stability of polypeptides throughout their life cycle remains a challenge. Electrohydrodynamic (EHD) techniques have been widely explored for encapsulation and delivery of many biopharmaceuticals. In this work, we explored monoaxial electrospraying for encapsulation of bovine liver catalase, investigating the effects of the different components of the electrospraying solution on the integrity and bioactivity of the enzyme. The catalase was successfully encapsulated within polymeric particles made of polyvinylpyrrolidone (PVP), dextran, and polysucrose. The polysorbate 20 content within the electrospraying solution (50 mM citrate buffer, pH 5.4) affected the catalase loading—increasing the polysorbate 20 concentration to 500 μg/mL resulted in full protein encapsulation but did not prevent loss in activity. The addition of ethanol (20% v/v) to a fully aqueous solution improves the electrospraying process by reducing surface tension, without loss of catalase activity. The polymer type was shown to have the greatest impact on preserving catalase activity within the electrosprayed particles. When PVP was the carrier there was no loss in activity compared with fresh aqueous solutions of catalase. The optimum particles were obtained from a 20% w/v PVP or 30% w/v PVP-trehalose (1:1 w/w) solution. The addition of trehalose confers stability advantages to the catalase particles. When trehalose-PVP particles were stored at 5 °C, enzymatic activity was maintained over 3 months, whereas for the PVP-only analogue a 50% reduction in activity was seen. This demonstrates that processing catalase by monoaxial electrospraying can, under optimised conditions, result in stable polymeric particles with no loss of activity.
Collapse
|
125
|
Arib C, Griveau A, Eyer J, Spadavecchia J. Cell penetrating peptide (CPP) gold(iii) - complex - bioconjugates: from chemical design to interaction with cancer cells for nanomedicine applications. NANOSCALE ADVANCES 2022; 4:3010-3022. [PMID: 36133522 PMCID: PMC9417459 DOI: 10.1039/d2na00096b] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/11/2022] [Indexed: 05/14/2023]
Abstract
This study promotes an innovative synthesis of a nanotheragnostic scaffold capable of targeting and destroying pancreatic cancer cells (PDAC) using the Biotinylated NFL-TBS.40-63 peptide (BIOT-NFL), known to enter various glioblastoma cancer cells (GBM) where it specifically destroys their microtubule network. This recently proposed methodology (P7391FR00-50481 LIV) applied to other peptides VIM (Vimentin) and TAT (Twin-Arginine Translocation) (CPP peptides) has many advantages, such as targeted selective internalization and high stability under experimental conditions, modulated by steric and chemical configurations of peptides. The successful interaction of peptides on gold surfaces has been confirmed by UV-visible, dynamic light scattering (DLS), Zeta potential (ZP) and Raman spectroscopy analyses. The cellular internalization in pancreatic ductal adenocarcinoma (PDAC; MIA PACA-2) and GBM (F98) cells was monitored by transmission electron microscopy (TEM) and showed a better cellular internalization in the presence of peptides with gold nanoparticles. In this work, we also evaluated the power of these hybrid peptide-nanoparticles as photothermal agents after cancer cell internalization. These findings envisage novel perspectives for the development of high peptide-nanotheragnostics.
Collapse
Affiliation(s)
- Celia Arib
- CNRS, UMR 7244, CSPBAT, Laboratoire de Chimie, Structures et Propriétés de Biomatériaux et d'Agents Thérapeutiques Université Paris 13 Sorbonne Paris Cité Bobigny France
| | - Audrey Griveau
- Laboratoire Micro et Nanomedecines Translationnelles, Inserm 1066, CNRS 6021, Institut de Recherche en Ingénierie de la Sante, Bâtiment IBS Institut de Biologie de la Sante, Université, Angers, Centre Hospitalier Universitaire Angers France
| | - Joel Eyer
- Laboratoire Micro et Nanomedecines Translationnelles, Inserm 1066, CNRS 6021, Institut de Recherche en Ingénierie de la Sante, Bâtiment IBS Institut de Biologie de la Sante, Université, Angers, Centre Hospitalier Universitaire Angers France
| | - Jolanda Spadavecchia
- CNRS, UMR 7244, CSPBAT, Laboratoire de Chimie, Structures et Propriétés de Biomatériaux et d'Agents Thérapeutiques Université Paris 13 Sorbonne Paris Cité Bobigny France
| |
Collapse
|
126
|
Gibson K, Cooper-Shepherd DA, Pallister E, Inman SE, Jackson SE, Lindo V. Toward Rapid Aspartic Acid Isomer Localization in Therapeutic Peptides Using Cyclic Ion Mobility Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2022; 33:1204-1212. [PMID: 35609180 PMCID: PMC9264384 DOI: 10.1021/jasms.2c00053] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
There is an increasing emphasis on the critical evaluation of interbatch purity and physical stability of therapeutic peptides. This is due to concerns over the impact that product- and process-related impurities may have on safety and efficacy of this class of drug. Aspartic acid isomerization to isoaspartic acid is a common isobaric impurity that can be very difficult to identify without first synthesizing isoAsp peptide standards for comparison by chromatography. As such, analytical tools that can determine if an Asp residue has isomerized, as well as the site of isomerization within the peptide sequence, are highly sought after. Ion mobility-mass spectrometry is a conformation-selective method that has developed rapidly in recent years particularly with the commercialization of traveling wave ion mobility instruments. This study employed a cyclic ion mobility (cIMS) mass spectrometry system to investigate the conformational characteristics of a therapeutic peptide and three synthetic isomeric forms, each with a single Asp residue isomerized to isoAsp. cIMS was able to not only show distinct conformational differences between each peptide but crucially, in conjunction with a simple workflow for comparing ion mobility data, it correctly located which Asp residue in each peptide had isomerized to isoAsp. This work highlights the value of cIMS as a potential screening tool in the analysis of therapeutic peptides prone to the formation of isoAsp impurities.
Collapse
Affiliation(s)
- Katherine Gibson
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
- Analytical
Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge CB21 6GH, U.K.
| | | | - Edward Pallister
- Analytical
Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge CB21 6GH, U.K.
| | - Sophie E. Inman
- Analytical
Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge CB21 6GH, U.K.
| | - Sophie E. Jackson
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| | - Viv Lindo
- Analytical
Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge CB21 6GH, U.K.
| |
Collapse
|
127
|
Bojko M, Węgrzyn K, Sikorska E, Kocikowski M, Parys M, Battin C, Steinberger P, Kogut MM, Winnicki M, Sieradzan AK, Spodzieja M, Rodziewicz-Motowidło S. Design, synthesis and biological evaluation of PD-1 derived peptides as inhibitors of PD-1/PD-L1 complex formation for cancer therapy. Bioorg Chem 2022; 128:106047. [DOI: 10.1016/j.bioorg.2022.106047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/04/2022] [Accepted: 07/19/2022] [Indexed: 12/11/2022]
|
128
|
Sunny S, Jyothidasan A, David CL, Parsawar K, Veerappan A, Jones DP, Pogwizd S, Rajasekaran NS. Tandem Mass Tagging Based Identification of Proteome Signatures for Reductive Stress Cardiomyopathy. Front Cardiovasc Med 2022; 9:848045. [PMID: 35770227 PMCID: PMC9234166 DOI: 10.3389/fcvm.2022.848045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/14/2022] [Indexed: 12/15/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2), a redox sensor, is vital for cellular redox homeostasis. We reported that transgenic mice expressing constitutively active Nrf2 (CaNrf2-TG) exhibit reductive stress (RS). In this study, we identified novel protein signature for RS-induced cardiomyopathy using Tandem Mass Tag (TMT) proteomic analysis in heart tissues of TG (CaNrf2-TG) mice at 6–7 months of age. A total of 1,105 proteins were extracted from 22,544 spectra. About 560 proteins were differentially expressed in TG vs. NTg hearts, indicating a global impact of RS on the myocardial proteome. Over 32 proteins were significantly altered in response to RS -20 were upregulated and 12 were downregulated in the hearts of TG vs. NTg mice, suggesting that these proteins could be putative signatures of RS. Scaffold analysis revealed a clear distinction between TG vs. NTg hearts. The majority of the differentially expressed proteins (DEPs) that were significantly altered in RS mice were found to be involved in stress related pathways such as antioxidants, NADPH, protein quality control, etc. Interestingly, proteins that were involved in mitochondrial respiration, lipophagy and cardiac rhythm were dramatically decreased in TG hearts. Of note, we identified the glutathione family of proteins as the significantly changed subset of the proteome in TG heart. Surprisingly, our comparative analysis of NGS based transcriptome and TMT-proteome indicated that ~50% of the altered proteins in TG myocardium was found to be negatively correlated with their transcript levels. In association with the altered proteome the TG mice displayed pathological cardiac remodeling.
Collapse
Affiliation(s)
- Sini Sunny
- Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Arun Jyothidasan
- Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Cynthia L David
- Analytical and Biological Mass Spectrometry Core Facility, The University of Arizona, Tuscon, AZ, United States
| | - Krishna Parsawar
- Analytical and Biological Mass Spectrometry Core Facility, The University of Arizona, Tuscon, AZ, United States
| | - Arul Veerappan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, New York University School of Medicine, New York, NY, United States.,Department of Environmental Medicine, New York University School of Medicine, New York, NY, United States
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States
| | - Steven Pogwizd
- Comprehensive Cardiovascular Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Namakkal S Rajasekaran
- Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, United States.,Division of Cardiovascular Medicine, Department of Medicine, The University of Utah, Salt Lake City, UT, United States.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
129
|
Indrakumar S, Kulakova A, Harris P, Peters GHJ. Dynamics of Human Serum Transferrin in Varying Physicochemical Conditions Explored by Using Molecular Dynamics Simulations. Mol Pharm 2022; 19:2795-2806. [PMID: 35776490 DOI: 10.1021/acs.molpharmaceut.2c00158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Conformational stability of human serum transferrin (Tf) at varying pH values and salt and excipient concentrations were investigated using molecular dynamics (MD) simulations, and the results are compared with previously published small-angle X-ray scattering (SAXS) experiments. SAXS study showed that at pH 5, Tf is predominantly present in a partially open (PO) form, and the factions of PO differ based on the physicochemical condition and drift toward the closed form (HO) as the pH increases. Tf is a bilobal glycoprotein that is composed of homologous halves termed the N- and C-lobes. The current study shows that the protonation of Y188 and K206 at pH 5 is the primary conformational drive into PO, which shifts toward the closed (HO) conformer as the pH increases. Furthermore, at pH 6.5, PO is unfavorable due to negative charge-charge repulsion at the N/C-lobe interface linker region causing increased hinge distance when compared to HO, which has favorable attractive electrostatic interactions in this region. Subsequently, the effect of salt concentration was studied at 70 and 140 mM NaCl. At 70 mM NaCl and pH 5, chloride ions bind strongly in the N-lobe iron-binding site, whereas these interactions are weak at pH 6.5. With increasing salt concentration at pH 5, the regions surrounding the N-lobe iron-binding site are saturated, and as a consequence, sodium and chloride ions accumulate into the bulk. Additionally, protein-excipient interactions were investigated. At pH 5, the excipients interact in similar loop regions, E89-T93, and D416-D420, located in the N- and C-lobes of the HO conformer, respectively. It is anticipated that interactions of additives in these two loop regions cause conformational changes that lead to iron-coordinating residues in the N-lobe to drift away from iron and thus drive HO to PO conversion. Furthermore, at pH 6.5 and 140 mM histidine, these interactions are negligible leading to the stabilization of HO.
Collapse
Affiliation(s)
- Sowmya Indrakumar
- Technical University of Denmark, Department of Chemistry, 2800 Kgs. Lyngby, Denmark
| | - Alina Kulakova
- Technical University of Denmark, Department of Chemistry, 2800 Kgs. Lyngby, Denmark
| | - Pernille Harris
- Technical University of Denmark, Department of Chemistry, 2800 Kgs. Lyngby, Denmark
| | - Günther H J Peters
- Technical University of Denmark, Department of Chemistry, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
130
|
Czechtizky W, Su W, Ripa L, Schiesser S, Höijer A, Cox RJ. Advances in the design of new types of inhaled medicines. PROGRESS IN MEDICINAL CHEMISTRY 2022; 61:93-162. [PMID: 35753716 DOI: 10.1016/bs.pmch.2022.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Inhalation of small molecule drugs has proven very efficacious for the treatment of respiratory diseases due to enhanced efficacy and a favourable therapeutic index compared with other dosing routes. It enables targeted delivery to the lung with rapid onset of therapeutic action, low systemic drug exposure, and thereby reduced systemic side effects. An increasing number of pharmaceutical companies and biotechs are investing in new modalities-for this review defined as therapeutic molecules with a molecular weight >800Da and therefore beyond usual inhaled small molecule drug-like space. However, our experience with inhaled administration of PROTACs, peptides, oligonucleotides (antisense oligonucleotides, siRNAs, miRs and antagomirs), diverse protein scaffolds, antibodies and antibody fragments is still limited. Investigating the retention and metabolism of these types of molecules in lung tissue and fluid will contribute to understanding which are best suited for inhalation. Nonetheless, the first such therapeutic molecules have already reached the clinic. This review will provide information on the physiology of healthy and diseased lungs and their capacity for drug metabolism. It will outline the stability, aggregation and immunogenicity aspects of new modalities, as well as recap on formulation and delivery aspects. It concludes by summarising clinical trial outcomes with inhaled new modalities based on information available at the end of 2021.
Collapse
Affiliation(s)
- Werngard Czechtizky
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden.
| | - Wu Su
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Lena Ripa
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Stefan Schiesser
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Andreas Höijer
- Cardiovascular, Renal & Metabolism CMC Projects, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Rhona J Cox
- Department of Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal & Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| |
Collapse
|
131
|
Sun J, Liu X, Wang Z, Yin F, Liu H, Nakamura Y, Yu C, Zhou D. Gastrointestinal digestion and absorption characterization in vitro of zinc-chelating hydrolysate from scallop adductor (Patinopecten yessoensis). JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:3277-3286. [PMID: 34802153 DOI: 10.1002/jsfa.11673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 09/17/2021] [Accepted: 11/21/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUD Zinc (Zn) is an essential catalytic element in the human health system but its absorption in the intestinal system can be strongly affected by gastrointestinal (GI) digestion. In this study, the food-derived potential Zn carrier, scallop adductor hydrolysates (SAHs), was produced and characterized. RESULTS During temporary storage at 4 °C, SAH decreased in Zn-chelating capacity in the aqueous phase, whereas the SAH-Zn complex exhibited high stability. Moreover, the secondary structure of SAH had no significant alteration. Zn morphologically altered the surface structures of SAH, which was involving in carboxyl group of SAH. Results of in vitro GI digestion suggested that the SAH-Zn maintained good stability in GI system and only proportion of high molecular weight cleaved. In addition, SAH could successfully carry and transport Zn while the fluorescence staining revealed free Zn accumulation inside the tissue. Finally, three representative absorbed peptides (around 600 Da) were identified and synthesized. Three synthetic peptides exhibit higher Zn-chelating capacity than SAH and could also successfully transported through the intestine. CONCLUSION This study provided a theoretical basis for the investigation of digestion and absorption of marine animal-derived peptides as Zn carriers. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiatong Sun
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Xiaoyang Liu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National Engineering Research Center of Seafood, Dalian, China
| | - Zixu Wang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Fawen Yin
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National Engineering Research Center of Seafood, Dalian, China
| | - Huilin Liu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National Engineering Research Center of Seafood, Dalian, China
| | - Yoshimasa Nakamura
- Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Chenxu Yu
- National Engineering Research Center of Seafood, Dalian, China
- Department of Agricultural and Biosystems Engineering, Iowa State University, Ames, IA, USA
| | - Dayong Zhou
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National Engineering Research Center of Seafood, Dalian, China
| |
Collapse
|
132
|
Sharma N, Kukreja D, Giri T, Kumar S, Shah RP. Synthetic pharmaceutical peptides characterization by chromatography principles and method development. J Sep Sci 2022; 45:2200-2216. [PMID: 35460196 DOI: 10.1002/jssc.202101034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 11/08/2022]
Abstract
As per United States Food and Drug Administration, any polymer/chain composed of 40 or fewer amino acids is called as a peptide, where more than 40 amino acids are considered as proteins. In many occasions there is a change in the source of manufacturing of the peptide active pharmaceutical ingredient, where one has to prove the sameness of that product with the existing formulation by considering several aspects like presence of impurities/degradation products, extent of aggregations etc. For the same, several chromatographic characterization techniques such as; Reverse phase high performance liquid chromatography-ultraviolet/high resolution mass spectrometry, supercritical fluid chromatography, size exclusion chromatography, Ion exchange chromatography etc are widely used in pharmaceutical industry. It is well known that the method development of peptide molecules is often challenging as many variables are to be kept in mind which can affect the separation, recovery and stability of molecule. The present review focuses on the basics of peptide degradation and method development by using various chromatographic techniques for characterization. It also covers a deep insight of method development parameters and variables to be considered which might directly or indirectly affect the chromatographic separation and recovery, and also provides a guide on selection of chromatographic parameters. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nitish Sharma
- The National Institute of Pharmaceutical Education and Research-Ahmedabad (Ministry of Chemicals and Fertilizers Government of India), Opposite Air force Station, Palaj, Gandhinagar, Gujarat, 382355, INDIA
| | - Divya Kukreja
- The National Institute of Pharmaceutical Education and Research-Ahmedabad (Ministry of Chemicals and Fertilizers Government of India), Opposite Air force Station, Palaj, Gandhinagar, Gujarat, 382355, INDIA
| | - Tushar Giri
- The National Institute of Pharmaceutical Education and Research-Ahmedabad (Ministry of Chemicals and Fertilizers Government of India), Opposite Air force Station, Palaj, Gandhinagar, Gujarat, 382355, INDIA
| | - Sumit Kumar
- The National Institute of Pharmaceutical Education and Research-Ahmedabad (Ministry of Chemicals and Fertilizers Government of India), Opposite Air force Station, Palaj, Gandhinagar, Gujarat, 382355, INDIA
| | - Ravi P Shah
- The National Institute of Pharmaceutical Education and Research-Ahmedabad (Ministry of Chemicals and Fertilizers Government of India), Opposite Air force Station, Palaj, Gandhinagar, Gujarat, 382355, INDIA
| |
Collapse
|
133
|
Kuncewicz K, Battin C, Węgrzyn K, Sieradzan A, Wardowska A, Sikorska E, Giedrojć I, Smardz P, Pikuła M, Steinberger P, Rodziewicz-Motowidło S, Spodzieja M. Targeting the HVEM protein using a fragment of glycoprotein D to inhibit formation of the BTLA/HVEM complex. Bioorg Chem 2022; 122:105748. [PMID: 35325694 DOI: 10.1016/j.bioorg.2022.105748] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/26/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy using blockade of immune checkpoints is mainly based on monoclonal antibodies. Despite the tremendous success achieved by using those molecules to block immune checkpoint proteins, antibodies possess some weaknesses, which means that there is still a need to search for new compounds as alternatives to antibodies. Many current approaches are focused on use of peptides/peptidomimetics to destroy receptor/ligand interactions. Our studies concern blockade of the BTLA/HVEM complex, which generates an inhibitory effect on the immune response resulting in tolerance to cancer cells. To design inhibitors of such proteins binding we based our work on the amino acid sequence and structure of a ligand of HVEM protein, namely glycoprotein D, which possesses the same binding site on HVEM as BTLA protein. To disrupt the BTLA and HVEM interaction we designed several peptides, all fragments of glycoprotein D, and tested their binding to HVEM using SPR and their ability to inhibit the BTLA/HVEM complex formation using ELISA tests and cellular reporter platforms. That led to identification of two peptides, namely gD(1-36)(K10C-D30C) and gD(1-36)(A12C-L25C), which interact with HVEM and possess blocking capacities. Both peptides are not cytotoxic to human PBMCs, and show stability in human plasma. We also studied the 3D structure of the gD(1-36)(K10C-D30C) peptide using NMR and molecular modeling methods. The obtained data reveal that it possesses an unstructured conformation and binds to HVEM in the same location as gD and BTLA. All these results suggest that peptides based on the binding fragment of gD protein represent promising immunomodulation agents for future cancer immunotherapy.
Collapse
Affiliation(s)
| | - Claire Battin
- Medical University of Vienna, Institute of Immunology, Division of Immune Receptors and T cell Activation, 1090 Vienna, Austria
| | - Katarzyna Węgrzyn
- University of Gdańsk, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, 80-307 Gdańsk, Poland
| | - Adam Sieradzan
- University of Gdańsk, Faculty of Chemistry, 80-308 Gdańsk, Poland
| | - Anna Wardowska
- Medical University of Gdańsk, Department of Physiopathology, 80-210 Gdańsk, Poland
| | - Emilia Sikorska
- University of Gdańsk, Faculty of Chemistry, 80-308 Gdańsk, Poland
| | - Irma Giedrojć
- University of Gdańsk, Faculty of Chemistry, 80-308 Gdańsk, Poland
| | - Pamela Smardz
- University of Gdańsk, Faculty of Chemistry, 80-308 Gdańsk, Poland
| | - Michał Pikuła
- Medical University of Gdańsk, Department of Embryology, Laboratory of Tissue Engineering and Regenerative Medicine, 80-210 Gdańsk, Poland
| | - Peter Steinberger
- Medical University of Vienna, Institute of Immunology, Division of Immune Receptors and T cell Activation, 1090 Vienna, Austria
| | | | - Marta Spodzieja
- University of Gdańsk, Faculty of Chemistry, 80-308 Gdańsk, Poland.
| |
Collapse
|
134
|
Hjalte J, Hossain S, Hugerth A, Sjögren H, Wahlgren M, Larsson P, Lundberg D. Aggregation Behavior of Structurally Similar Therapeutic Peptides Investigated by 1H NMR and All-Atom Molecular Dynamics Simulations. Mol Pharm 2022; 19:904-917. [PMID: 35104408 PMCID: PMC8905580 DOI: 10.1021/acs.molpharmaceut.1c00883] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 11/30/2022]
Abstract
Understanding of peptide aggregation propensity is an important aspect in pharmaceutical development of peptide drugs. In this work, methodologies based on all-atom molecular dynamics (AA-MD) simulations and 1H NMR (in neat H2O) were evaluated as tools for identification and investigation of peptide aggregation. A series of structurally similar, pharmaceutically relevant peptides with known differences in aggregation behavior (D-Phe6-GnRH, ozarelix, cetrorelix, and degarelix) were investigated. The 1H NMR methodology was used to systematically investigate variations in aggregation with peptide concentration and time. Results show that 1H NMR can be used to detect the presence of coexisting classes of aggregates and the inclusion or exclusion of counterions in peptide aggregates. Interestingly, results suggest that the acetate counterions are included in aggregates of ozarelix and cetrorelix but not in aggregates of degarelix. The peptides investigated in AA-MD simulations (D-Phe6-GnRH, ozarelix, and cetrorelix) showed the same rank order of aggregation propensity as in the NMR experiments. The AA-MD simulations also provided molecular-level insights into aggregation dynamics, aggregation pathways, and the influence of different structural elements on peptide aggregation propensity and intermolecular interactions within the aggregates. Taken together, the findings from this study illustrate that 1H NMR and AA-MD simulations can be useful, complementary tools in early evaluation of aggregation propensity and formulation development for peptide drugs.
Collapse
Affiliation(s)
- Johanna Hjalte
- Food
Technology, Engineering and Nutrition, Lund
University, Box 124, 221 00 Lund, Sweden
| | - Shakhawath Hossain
- Department
of Pharmacy, Drug Delivery, Uppsala University, Box 580, 751 23 Uppsala, Sweden
| | - Andreas Hugerth
- Ferring
Pharmaceuticals A/S, Amager Strandvej 405, 2770 Kastrup, Denmark
| | - Helen Sjögren
- Ferring
Pharmaceuticals A/S, Amager Strandvej 405, 2770 Kastrup, Denmark
| | - Marie Wahlgren
- Food
Technology, Engineering and Nutrition, Lund
University, Box 124, 221 00 Lund, Sweden
| | - Per Larsson
- Department
of Pharmacy, Drug Delivery, Uppsala University, Box 580, 751 23 Uppsala, Sweden
| | - Dan Lundberg
- CR
Competence AB, Center for Chemistry and Chemical Engineering, Box 124, 221 00 Lund, Sweden
| |
Collapse
|
135
|
Castelletto V, Hamley IW. Amyloid
and Hydrogel Formation of a Peptide Sequence
from a Coronavirus Spike Protein. ACS NANO 2022; 16:1857-1867. [PMCID: PMC8867915 DOI: 10.1021/acsnano.1c10658] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/29/2021] [Indexed: 05/28/2023]
Abstract
![]()
We demonstrate that
a conserved coronavirus spike protein peptide
forms amyloid structures, differing from the native helical conformation
and not predicted by amyloid aggregation algorithms. We investigate
the conformation and aggregation of peptide RSAIEDLLFDKV,
which is a sequence common to many animal and human coronavirus spike
proteins. This sequence is part of a native α-helical S2 glycoprotein
domain, close to and partly spanning the fusion sequence. This peptide
aggregates into β-sheet amyloid nanotape structures close to
the calculated pI = 4.2, but forms disordered monomers at high and
low pH. The β-sheet conformation revealed by FTIR and circular
dichroism (CD) spectroscopy leads to peptide nanotape structures,
imaged using transmission electron microscopy (TEM) and probed by
small-angle X-ray scattering (SAXS). The nanotapes comprise arginine-coated
bilayers. A Congo red dye UV–vis assay is used to probe the
aggregation of the peptide into amyloid structures, which enabled
the determination of a critical aggregation concentration (CAC). This
peptide also forms hydrogels under precisely defined conditions of
pH and concentration, the rheological properties of which were probed.
The observation of amyloid formation by a coronavirus spike has relevance
to the stability of the spike protein conformation (or its destabilization via pH change), and the peptide may have potential utility
as a functional material. Hydrogels formed by coronavirus peptides
may also be of future interest in the development of slow-release
systems, among other applications.
Collapse
Affiliation(s)
- Valeria Castelletto
- Department
of Chemistry, University of Reading, Reading RG6 6AD, United Kingdom
| | - Ian W. Hamley
- Department
of Chemistry, University of Reading, Reading RG6 6AD, United Kingdom
| |
Collapse
|
136
|
Yu A, Dai X, Wang Z, Chen H, Guo B, Huang L. Recent Advances of Mesoporous Silica as a Platform for Cancer Immunotherapy. BIOSENSORS 2022; 12:109. [PMID: 35200369 PMCID: PMC8869707 DOI: 10.3390/bios12020109] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 05/06/2023]
Abstract
Immunotherapy is a promising modality of treatment for cancer. Immunotherapy is comprised of systemic and local treatments that induce an immune response, allowing the body to fight back against cancer. Systemic treatments such as cancer vaccines harness antigen presenting cells (APCs) to activate T cells with tumor-associated antigens. Small molecule inhibitors can be employed to inhibit immune checkpoints, disrupting tumor immunosuppression and immune evasion. Despite the current efficacy of immunotherapy, improvements to delivery can be made. Nanomaterials such as mesoporous silica can facilitate the advancement of immunotherapy. Mesoporous silica has high porosity, decent biocompatibility, and simple surface functionalization. Mesoporous silica can be utilized as a versatile carrier of various immunotherapeutic agents. This review gives an introduction on mesoporous silica as a nanomaterial, briefly covering synthesis and biocompatibility, and then an overview of the recent progress made in the application of mesoporous silica to cancer immunotherapy.
Collapse
Affiliation(s)
- Albert Yu
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Xiaoyong Dai
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Zixian Wang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Huaqing Chen
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Bing Guo
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China;
| | - Laiqiang Huang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
137
|
Basak A, Basak S. Protein Aggregation and Self Assembly in Health and Disease. CURR PROTEOMICS 2022. [DOI: 10.2174/1570164618666210223160742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Self-attachment of proteins leading to the formation of highly insoluble protein oligomers and aggregates has become an important focus of research owing to its diverse implications in pathophysiology and diseases. This has become a more frequent phenomenon in most neurological and neurodegenerative diseases as well as in dementia. In recent years such event of protein aggregation has linked to other disease conditions, disorders or adverse health conditions. Interestingly, aggregation of protein also plays role in development, growth or metabolism. Most often physiological proteins are initially bio-synthesised in native or nascent geometrical forms or conformations but later they undergo specific folding pattern and thereby acquire a stable configuration that is biologically relevant and active. It is highly important that these proteins remain in their biologically active configuration in order to exert their functional properties. Any alteration or change to this structural configuration can be detrimental to their specific functions and may cause pathological consequences leading to the onset of diseases or disorders. Several factors such as the action of chaperones, binding partners, physiological metal ions, pH level, temperature, ionic strength, interfacial exposure (solid-liquid, liquid-liquid, gas-liquid), mutation and post translational modification, chemical changes, interaction with small molecules such as lipids, hormones, etc. and solvent environment have been either identified or proposed as important factors in conferring the ultimate status of protein structure and configuration.
Among many misfolding protein conformations, self-assembly or aggregation is the most significant. It leads to the formation of highly oligomeric self-aggregates that precipitate and interfere with many biochemical processes with serious pathological consequences. The most common implication of protein aggregation leading to the formation of deposits / plaques of various morphological types is the onset of neurological and neurodegenerative diseases that include Alzheimer’s, Parkinson’s, Huntington, ALS (Amyotrophic Lateral Sclerosis), CJD (Creutzfeldt Jakob Dementia), Prion diseases, Amyloidosis and other forms of dementia. However increasingly studies revealed that protein aggregation may also be associated with other diseases such as cancer, type 2 diabetes, renal, corneal and cardiovascular diseases. Protein aggregation diseases are now considered as part of “Proteinopathy” which refers to conditions where proteins become structurally abnormal or fail to fold into stable normal configurations. In this review, we reflect on various aspects of protein self-aggregation, potential underlying causes, mechanism, role of secondary structures, pathological consequences and possible intervention strategies as reported in published literatures.
Collapse
Affiliation(s)
- Ajoy Basak
- Pathology and Laboratory Medicine, Faculty of Medicine, U Ottawa, Canada
- Ottawa Hospital Research Institute,
The Ottawa Hospital, U Ottawa, Canada
| | - Sarmistha Basak
- Formerly of Kidney Research Center, Ottawa Hospital Research Institute, U Ottawa, Canada
| |
Collapse
|
138
|
Gatto E, Toniolo C, Venanzi M. Peptide Self-Assembled Nanostructures: From Models to Therapeutic Peptides. NANOMATERIALS 2022; 12:nano12030466. [PMID: 35159810 PMCID: PMC8838750 DOI: 10.3390/nano12030466] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/25/2022]
Abstract
Self-assembly is the most suitable approach to obtaining peptide-based materials on the nano- and mesoscopic scales. Applications span from peptide drugs for personalized therapy to light harvesting and electron conductive media for solar energy production and bioelectronics, respectively. In this study, we will discuss the self-assembly of selected model and bioactive peptides, in particular reviewing our recent work on the formation of peptide architectures of nano- and mesoscopic size in solution and on solid substrates. The hierarchical and cooperative characters of peptide self-assembly will be highlighted, focusing on the structural and dynamical properties of the peptide building blocks and on the nature of the intermolecular interactions driving the aggregation phenomena in a given environment. These results will pave the way for the understanding of the still-debated mechanism of action of an antimicrobial peptide (trichogin GA IV) and the pharmacokinetic properties of a peptide drug (semaglutide) currently in use for the therapy of type-II diabetes.
Collapse
Affiliation(s)
- Emanuela Gatto
- PEPSA-LAB, Department of Chemical Science and Technologies, University of Rome, Tor Vergata, 00133 Rome, Italy;
| | - Claudio Toniolo
- Department of Chemical Sciences, University of Padua, 35131 Padua, Italy;
| | - Mariano Venanzi
- PEPSA-LAB, Department of Chemical Science and Technologies, University of Rome, Tor Vergata, 00133 Rome, Italy;
- Correspondence: ; Tel.: +39-06-7259-4468
| |
Collapse
|
139
|
Evidence for a semisolid phase state of aerosols and droplets relevant to the airborne and surface survival of pathogens. Proc Natl Acad Sci U S A 2022; 119:2109750119. [PMID: 35064080 PMCID: PMC8794803 DOI: 10.1073/pnas.2109750119] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2021] [Indexed: 01/25/2023] Open
Abstract
Ambient humidity can influence the survival of pathogens in respiratory aerosols and droplets, although the mechanism and optimum humidity level for public health remain unclear. Here, we present evidence for a humidity-dependent, semisolid state of aerosols and droplets relevant to pathogen survival. These observations indicate that a semisolid state may protect pathogens from inactivation by hindering disinfection reactions at intermediate-to-low humidity levels. The formation of the semisolid state was dependent on the composition of the aerosols, which suggests that the humidity for optimum pathogen destruction will depend on the composition of respiratory particles released from an infected host. These observations can be used to help interpret laboratory studies and inform public health recommendations. The phase state of respiratory aerosols and droplets has been linked to the humidity-dependent survival of pathogens such as SARS-CoV-2. To inform strategies to mitigate the spread of infectious disease, it is thus necessary to understand the humidity-dependent phase changes associated with the particles in which pathogens are suspended. Here, we study phase changes of levitated aerosols and droplets composed of model respiratory compounds (salt and protein) and growth media (organic–inorganic mixtures commonly used in studies of pathogen survival) with decreasing relative humidity (RH). Efflorescence was suppressed in many particle compositions and thus unlikely to fully account for the humidity-dependent survival of viruses. Rather, we identify organic-based, semisolid phase states that form under equilibrium conditions at intermediate RH (45 to 80%). A higher-protein content causes particles to exist in a semisolid state under a wider range of RH conditions. Diffusion and, thus, disinfection kinetics are expected to be inhibited in these semisolid states. These observations suggest that organic-based, semisolid states are an important consideration to account for the recovery of virus viability at low RH observed in previous studies. We propose a mechanism in which the semisolid phase shields pathogens from inactivation by hindering the diffusion of solutes. This suggests that the exogenous lifetime of pathogens will depend, in part, on the organic composition of the carrier respiratory particle and thus its origin in the respiratory tract. Furthermore, this work highlights the importance of accounting for spatial heterogeneities and time-dependent changes in the properties of aerosols and droplets undergoing evaporation in studies of pathogen viability.
Collapse
|
140
|
Sen S, Ali R, Onkar A, Ganesh S, Verma S. Strategies for interference of insulin fibrillogenesis: challenges and advances. Chembiochem 2022; 23:e202100678. [PMID: 35025120 DOI: 10.1002/cbic.202100678] [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: 12/13/2021] [Revised: 01/11/2022] [Indexed: 11/10/2022]
Abstract
The discovery of insulin came up with very high hopes for diabetic patients. In the year 2021, the world celebrated the 100 th anniversary of the discovery of this vital hormone. However, external use of insulin is highly affected by its aggregating tendency that occurs during its manufacturing, transportation, and improper handling which ultimately leads its pharmaceutically and biologically ineffective form. In this review, we aim to discuss the various approaches used for decelerating insulin aggregation which results in the enhancement of its overall structural stability and usage. The approaches that are discussed are broadly classified as either a measure through excipient additions or by intrinsic modifications in the insulin native structure.
Collapse
Affiliation(s)
- Shantanu Sen
- Indian Institute of Technology Kanpur, Chemistry, INDIA
| | - Rafat Ali
- Indian Institute of Technology Kanpur, Chemistry, Room No 131 Lab No2, CESE department IIT Kanpur, 208016, Kanpur, INDIA
| | - Akanksha Onkar
- Indian Institute of Technology Kanpur, Biological Sciences and Bioengineering, INDIA
| | - Subramaniam Ganesh
- Indian Institute of Technology Kanpur, Biological Sciences and Bioengineering, INDIA
| | - Sandeep Verma
- Indian Institute of Technology-Kanpur, Department of Chemistry, IIT-Kanpur, 208016, Kanpur, INDIA
| |
Collapse
|
141
|
Grafting MSI-78A onto chitosan microspheres enhances its antimicrobial activity. Acta Biomater 2022; 137:186-198. [PMID: 34634508 DOI: 10.1016/j.actbio.2021.09.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022]
Abstract
MSI-78A (Pexiganan A) is one of the few antimicrobial peptides (AMPs) able to kill Helicobacter pylori, a pathogenic bacterium that colonizes the gastric mucosa of half of the world's population. Antibiotics fail in 20-40% of H. pylori-infected patients, reinforcing the need for alternative treatments. Herein, a bioengineered approach was developed. MSI-78A with a C-terminal cysteine was grafted onto chitosan microspheres (AMP-ChMic) by thiol-maleimide (Michael-addition) chemistry using a long heterobifunctional spacer (NHS-PEG113-MAL). Microspheres with ∼4 µm diameter (near H. pylori length) and stable at low pH were produced by spray drying using a chitosan solution with an incomplete genipin crosslinking. A 3 × 10-5 µg AMP/microsphere grafting was estimated/confirmed by UV/Vis and FTIR spectroscopies. AMP-ChMic were bactericidal against H. pylori J99 (highly pathogenic human strain) at lower concentrations than the free peptide (∼277 µg grafted MSI-78A-SH/mL vs 512 µg free MSI-78A-SH/mL), even after pre-incubation in simulated gastric conditions with pepsin. AMP-ChMic killed H. pylori by membrane destabilization and cytoplasm release in a ratio of ∼10 bacteria/microsphere. This can be attributed to H. pylori attraction to chitosan, facilitating the interaction of grafted AMP with bacterium membrane. Overall, it was demonstrated that the peptide-microsphere conjugation chemistry did not compromise the MSI-78A antimicrobial activity, instead it boosted its bactericidal performance against H. pylori. STATEMENT OF SIGNIFICANCE: Half of the world's population is infected with Helicobacter pylori, a gastric bacterium that is responsible for 90% of non-cardia gastric cancers. Therefore, H. pylori eradication is now advocated in all infected individuals. However, available antibiotic therapies fail in up to 40% patients. Antimicrobial peptides (AMPs) are appealing alternatives to antibiotics, but their high susceptibility in vivo limits their clinical translation. AMP immobilization onto biomaterials surface will overcome this problem. Herein, we demonstrate that immobilization of MSI-78A (one of the few AMPs with activity against H. pylori) onto chitosan microspheres (AMP-ChMic) enhances its anti-H. pylori activity even at acidic pH (gastric settings). These results highlight the strong potential of AMP-ChMic as an antibiotic alternative for H. pylori eradication.
Collapse
|
142
|
Jakaria SM, Budil DE, Murtagh J. Glycopeptide antibiotic drug stability in aqueous solution. AAPS OPEN 2022; 8:20. [PMCID: PMC9742044 DOI: 10.1186/s41120-022-00067-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/11/2022] [Indexed: 12/14/2022] Open
Abstract
Glycopeptide antimicrobials are a class of naturally occurring or semi-synthetic glycosylated products that have shown antibacterial activity against gram-positive organisms by inhibiting cell-wall synthesis. In most cases, these drugs are prepared in dry powder (lyophilized) form due to chemical and physical instability in aqueous solution; however, from an economic and practical point of view, liquid formulations are preferred. Researchers have recently found ways to formulate some glycopeptide antibiotic therapeutic drugs in aqueous solution at refrigerated or room temperature. Chemical degradation can be significantly slowed by formulating them at a defined pH with specific buffers, avoiding oxygen reactive species, and minimizing solvent exposure. Sugars, amino acids, polyols, and surfactants can reduce physical degradation by restricting glycopeptide mobility and reducing solvent interaction. This review focuses on recent studies on glycopeptide antibiotic drug stability in aqueous solution. It is organized into three sections: (i) glycopeptide antibiotic instability due to chemical and physical degradation, (ii) strategies to improve glycopeptide antibiotic stability in aqueous solution, and (iii) a survey of glycopeptide antibiotic drugs currently available in the market and their stability based on published literature and patents. Antimicrobial resistance deaths are expected to increase by 2050, making heat-stable glycopeptides in aqueous solution an important treatment option for multidrug-resistant and extensively drug-resistant pathogens. In conclusion, it should be possible to formulate heat stable glycopeptide drugs in aqueous solution by understanding the degradation mechanisms of this class of therapeutic drugs in greater detail, making them easily accessible to developing countries with a lack of cold chains.
Collapse
Affiliation(s)
- Sardar M. Jakaria
- Hikma Pharmaceuticals, Bedford, OH 44146 USA ,grid.261112.70000 0001 2173 3359Department of Chemistry and Chemical Biology, Northeastern University, MA 02115 Boston, USA
| | - David E. Budil
- grid.261112.70000 0001 2173 3359Department of Chemistry and Chemical Biology, Northeastern University, MA 02115 Boston, USA
| | | |
Collapse
|
143
|
Zhang F, Richter G, Bourgeois B, Spreitzer E, Moser A, Keilbach A, Kotnik P, Madl T. A General Small-Angle X-ray Scattering-Based Screening Protocol for Studying Physical Stability of Protein Formulations. Pharmaceutics 2021; 14:69. [PMID: 35056965 PMCID: PMC8778066 DOI: 10.3390/pharmaceutics14010069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
A fundamental step in developing a protein drug is the selection of a stable storage formulation that ensures efficacy of the drug and inhibits physiochemical degradation or aggregation. Here, we designed and evaluated a general workflow for screening of protein formulations based on small-angle X-ray scattering (SAXS). Our SAXS pipeline combines automated sample handling, temperature control, and fast data analysis and provides protein particle interaction information. SAXS, together with different methods including turbidity analysis, dynamic light scattering (DLS), and SDS-PAGE measurements, were used to obtain different parameters to provide high throughput screenings. Using a set of model proteins and biopharmaceuticals, we show that SAXS is complementary to dynamic light scattering (DLS), which is widely used in biopharmaceutical research and industry. We found that, compared to DLS, SAXS can provide a more sensitive measure for protein particle interactions, such as protein aggregation and repulsion. Moreover, we show that SAXS is compatible with a broader range of buffers, excipients, and protein concentrations and that in situ SAXS provides a sensitive measure for long-term protein stability. This workflow can enable future high-throughput analysis of proteins and biopharmaceuticals and can be integrated with well-established complementary physicochemical analysis pipelines in (biopharmaceutical) research and industry.
Collapse
Affiliation(s)
- Fangrong Zhang
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China;
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (G.R.); (B.B.); (E.S.)
| | - Gesa Richter
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (G.R.); (B.B.); (E.S.)
| | - Benjamin Bourgeois
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (G.R.); (B.B.); (E.S.)
| | - Emil Spreitzer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (G.R.); (B.B.); (E.S.)
| | - Armin Moser
- Anton Paar GmbH, 8054 Graz, Austria; (A.M.); (A.K.); (P.K.)
| | | | - Petra Kotnik
- Anton Paar GmbH, 8054 Graz, Austria; (A.M.); (A.K.); (P.K.)
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (G.R.); (B.B.); (E.S.)
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
144
|
Rajpoot S, Solanki K, Kumar A, Zhang KYJ, Pullamsetti SS, Savai R, Faisal SM, Pan Q, Baig MS. In-Silico Design of a Novel Tridecapeptide Targeting Spike Protein of SARS-CoV-2 Variants of Concern. Int J Pept Res Ther 2021; 28:28. [PMID: 34924897 PMCID: PMC8667532 DOI: 10.1007/s10989-021-10339-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2021] [Indexed: 12/11/2022]
Abstract
Several mutations in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have increased the transmission and mortality rate of coronavirus disease-19 (COVID-19) across the globe. Although many vaccines have been developed, a large proportion of the global population remains at high risk of infection. The current study aims to develop an antiviral peptide capable of inhibiting the interaction of SARS-CoV-2 spike protein and its six major variants with the host cell angiotensin-converting enzyme 2 (ACE2) receptor. An in-silico approach was employed to design a therapeutic peptide inhibitor against the receptor-binding domain (RBD) of the spike (S) protein of SARS-CoV-2 and its variants (B.1.1.7, B.1.351, P.1, B.1.617.1, B.1.617.2 and B.1.617.3). The binding specificity and affinity of our designed peptide inhibitor Mod13AApi (YADKYQKQYKDAY) with wild-type S-RBD and its six variants was confirmed by molecular docking using the HPEPDOCK tool, whereas complex stability was determined by the MD simulation study. The physicochemical and ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties of inhibitory peptides were determined using the ExPASy tool and pkCSM server. The docking results and its properties from our in-silico analysis present the Mod13AApi, a promising peptide for the rapid development of anti-coronavirus peptide-based antiviral therapy. Blockage of the binding of the spike protein of SARS-CoV-2 variants with ACE2 in the presence of the therapeutic peptide may prevent deadly SARS-CoV-2 variants entry into host cells. Therefore, the designed inhibitory peptide can be utilized as a promising therapeutic strategy to combat COVID-19, as evident from this in-silico study.
Collapse
Affiliation(s)
- Sajjan Rajpoot
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, 453552 India
| | - Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, 453552 India
| | - Ashutosh Kumar
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, Tsurumi, Yokohama, Kanagawa Japan
| | - Kam Y J Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, Tsurumi, Yokohama, Kanagawa Japan
| | - Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Hessen Germany.,Institute for Lung Health (ILH), Justus-Liebig-University Giessen, 35392 Giessen, Hessen Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Hessen Germany.,Institute for Lung Health (ILH), Justus-Liebig-University Giessen, 35392 Giessen, Hessen Germany
| | - Syed M Faisal
- National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana India
| | - Qiuwei Pan
- Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, 453552 India
| |
Collapse
|
145
|
Yadav A, Singh S, Sohi H, Dang S. Advances in Delivery of Chemotherapeutic Agents for Cancer Treatment. AAPS PharmSciTech 2021; 23:25. [PMID: 34907501 DOI: 10.1208/s12249-021-02174-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/29/2021] [Indexed: 12/31/2022] Open
Abstract
Presently, most of the treatment strategies for cancer are focused on the surgical removal of cancerous tumors, along with physical and chemical treatment such as radiotherapy and chemotherapy, respectively. The primary issue associated with these methods is the inhibition of normal cell growth and serious side effects associated with systemic toxicity. The traditional chemotherapeutics which were delivered systemically were inadequate and had serious dose limiting side effects. Recent advances in the development of chemotherapeutics have simultaneously paved the way for efficient targeted drug delivery. Despite the advances in the field of oncogenic drugs, several limitations remain, such as early blood clearance, acquired resistance against cytotoxic agents, toxicity associated with chemotherapeutics, and site-specific drug delivery. Hence, this review article focuses on the recent scientific advancements made in different types of drug delivery systems, including, organic nanocarriers (polymers, albumins, liposomes, and micelles), inorganic nanocarriers (mesoporous silica nanoparticles, gold nanoparticles, platinum nanoparticles, and carbon nanotubes), aptamers, antibody-drug conjugates, and peptides. These targeted drug delivery approaches offer numerous advantages such as site-specific drug delivery, minimal toxicity, better bioavailability, and an increased overall efficacy of the chemotherapeutics. Graphical abstract.
Collapse
|
146
|
César Moreira Brito J, Gustavo Lima W, Magalhães Resende J, Cristina Sampaio de Assis D, Boff D, Nascimento Cardoso V, Almeida Amaral F, Maria Souza-Fagundes E, Odília Antunes Fernandes S, Elena de Lima M. Pegylated LyeTx I-b peptide is effective against carbapenem-resistant Acinetobacter baumannii in an in vivo model of pneumonia and shows reduced toxicity. Int J Pharm 2021; 609:121156. [PMID: 34624440 DOI: 10.1016/j.ijpharm.2021.121156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/27/2021] [Accepted: 10/03/2021] [Indexed: 10/20/2022]
Abstract
The World Health Organization (WHO) has been warning about the importance of developing new drugs against superbugs. Antimicrobial peptides are an alternative in this context, most of them being involved in innate immunity, acting in various ways, and some even showing synergism with commercial antimicrobial agents. LyeTx I-b is a synthetic peptide derived from native LyeTx I, originally isolated from Lycosa erythrognatha spider venom. Although LyeTx I-b is active against several multidrug-resistant bacteria, it shows some hemolytic and cytotoxic effects. To overcome this hindrance, in the present study we PEGylated LyeTx I-b and evaluated its toxicity and in vitro and in vivo activities on pneumonia caused by multi-resistant Acinetobacter baumannii. PEGylated LyeTx I-b (LyeTx I-bPEG) maintained the same MIC value as the non- PEGylated peptide, showed anti-biofilm activity, synergistic effect with commercial antimicrobial agents, and did not induce resistance. Moreover, in vivo experiments showed its activity against pneumonia. Additionally, LyeTx I-bPEG reduced hemolysis up to 10 times, was approximately 2 times less cytotoxic to HEK-293 cells and 4 times less toxic to mice in acute toxicity models, compared to LyeTx I-b. Our results show LyeTx I-bPEG as a promising antimicrobial candidate, significantly active against pneumonia caused by multidrug-resistant A. baumannii.
Collapse
Affiliation(s)
- Júlio César Moreira Brito
- Programa de Inovação Tecnológica e Biofarmacêutica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil; Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil.
| | - William Gustavo Lima
- Laboratório de Radioisótopos, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus Pampulha, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jarbas Magalhães Resende
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Débora Cristina Sampaio de Assis
- Escola de Veterinária, Departamento de Inspeção Sanitária, Campus Pampulha, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Daiane Boff
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Valbert Nascimento Cardoso
- Laboratório de Radioisótopos, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus Pampulha, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flávio Almeida Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine Maria Souza-Fagundes
- Programa de Inovação Tecnológica e Biofarmacêutica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Simone Odília Antunes Fernandes
- Laboratório de Radioisótopos, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus Pampulha, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Elena de Lima
- Programa de Inovação Tecnológica e Biofarmacêutica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil; Faculdade Santa Casa de Belo Horizonte: Programa de Pós-Graduação em Medicina-Biomedicina, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
147
|
Armamentarium of Cryoprotectants in Peptide Vaccines: Mechanistic Insight, Challenges, Opportunities and Future Prospects. Int J Pept Res Ther 2021; 27:2965-2982. [PMID: 34690621 PMCID: PMC8524217 DOI: 10.1007/s10989-021-10303-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 10/30/2022]
Abstract
Vaccines are designed to leverage the immune system and produce long-lasting protection against specific diseases. Peptide vaccines are regarded as safe and effective way of circumventing problems such as mild allergic reactions associated with conventional vaccines. The biggest challenges associated with formulation of peptide vaccines are stability issues and conformational changes which lead to destruction of their activity when exposed to lyophilization process that may act as stressors. Lyophilization process is aimed at removal of water which involves freezing, primary drying and secondary drying. To safeguard the peptide molecules from such stresses, cryoprotectants are used to offer them viability and structural stability. This paper is an attempt to understand the physicochemical properties of peptide vaccines, mechanism of cryoprotection under the shed of water replacement, water substitution theory and cation-pi interaction theory of amino acids which aims at shielding the peptide from external environment by formation of hydrogen bonds, covalent bonds or cation-pi interaction between cryoprotectant and peptide followed by selection criteria of cryoprotectants and their utility in peptide vaccines development along with challenges and opportunities.
Collapse
|
148
|
Amyloidogenicity of peptides targeting diabetes and obesity. Colloids Surf B Biointerfaces 2021; 209:112157. [PMID: 34715595 DOI: 10.1016/j.colsurfb.2021.112157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/20/2021] [Accepted: 10/09/2021] [Indexed: 12/15/2022]
Abstract
Since the discovery of insulin, a century ago, the repertoire of therapeutic polypeptides targeting diabetes - and now also obesity - have increased substantially. The focus on quality has shifted from impure and unstable preparations of animal insulin to highly pure, homologous recombinant insulin, along with other peptide-based hormones and analogs such as amylin analogs (pramlintide, davalintide, cagrilintide), glucagon and glucagon-like peptide-1 receptor agonists (GLP-1, liraglutide, exenatide, semaglutide). Proper formulation, storage, manipulation and usage by professionals and patients are required in order to avoid agglomeration into high molecular weight products (HMWP), either amorphous or amyloid, which could result in potential loss of biological activity and short- or long-term immune reaction and silent inactivation. In this narrative review, we present perspective of the aggregation of therapeutic polypeptides used in diabetes and other metabolic diseases, covering the nature and mechanisms, analytical techniques, physical and chemical stability, strategies aimed to hamper the formation of HMWP, and perspectives on future biopharmaceutical developments.
Collapse
|
149
|
Noor A, Zafar S, Shafiq M, Younas N, Siegert A, Mann FA, Kruss S, Schmitz M, Dihazi H, Ferrer I, Zerr I. Molecular Profiles of Amyloid-β Proteoforms in Typical and Rapidly Progressive Alzheimer's Disease. Mol Neurobiol 2021; 59:17-34. [PMID: 34618331 PMCID: PMC8786784 DOI: 10.1007/s12035-021-02566-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/13/2021] [Indexed: 11/02/2022]
Abstract
The molecular determinants of atypical clinical variants of Alzheimer's disease, including the recently discovered rapidly progressive Alzheimer's disease (rpAD), are unknown to date. Fibrilization of the amyloid-β (Aβ) peptide is the most frequently studied candidate in this context. The Aβ peptide can exist as multiple proteoforms that vary in their post-translational processing, amyloidogenesis, and toxicity. The current study was designed to identify these variations in Alzheimer's disease patients exhibiting classical (sAD) and rapid progression, with the primary aim of establishing if these variants may constitute strains that underlie the phenotypic variability of Alzheimer's disease. We employed two-dimensional polyacrylamide gel electrophoresis and MALDI-ToF mass spectrometry to validate and identify the Aβ proteoforms extracted from targeted brain tissues. The biophysical analysis was conducted using RT-QuIC assay, confocal microscopy, and atomic force microscopy. Interactome analysis was performed by co-immunoprecipitation. We present a signature of 33 distinct pathophysiological proteoforms, including the commonly targeted Aβ40, Aβ42, Aβ4-42, Aβ11-42, and provide insight into their synthesis and quantities. Furthermore, we have validated the presence of highly hydrophobic Aβ seeds in rpAD brains that seeded reactions at a slower pace in comparison to typical Alzheimer's disease. In vitro and in vivo analyses also verified variations in the molecular pathways modulated by brain-derived Aβ. These variations in the presence, synthesis, folding, and interactions of Aβ among sAD and rpAD brains constitute important points of intervention. Further validation of reported targets and mechanisms will aid in the diagnosis of and therapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Aneeqa Noor
- Clinical Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Saima Zafar
- Clinical Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany. .,Biomedical Engineering and Sciences Department, School of Mechanical and Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Bolan Road, Islamabad, H-12, 44000, Pakistan.
| | - Mohsin Shafiq
- Clinical Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Neelam Younas
- Clinical Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Anna Siegert
- Clinical Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Florian A Mann
- Institute of Physical Chemistry, Georg-August University, Tammannstraße 6, 37077, Göttingen, Germany
| | - Sebastian Kruss
- Institute of Physical Chemistry, Georg-August University, Tammannstraße 6, 37077, Göttingen, Germany
| | - Matthias Schmitz
- Clinical Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Hassan Dihazi
- Department of Nephrology and Rheumatology, Georg-August University, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Isidre Ferrer
- Department of Pathology and Experimental Therapeutics, University of BarcelonaCIBERNEDBellvitge University Hospital (IDIBELL), Carrer de la Feixa Llarga, 08907, Hospitalet de Llobregat, Spain
| | - Inga Zerr
- Clinical Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| |
Collapse
|
150
|
Panchal D, Kataria J, Patel K, Crowe K, Pai V, Azizogli A, Kadian N, Sanyal S, Roy A, Dodd‐o J, Acevedo‐Jake AM, Kumar VA. Peptide-Based Inhibitors for SARS-CoV-2 and SARS-CoV. ADVANCED THERAPEUTICS 2021; 4:2100104. [PMID: 34514085 PMCID: PMC8420164 DOI: 10.1002/adtp.202100104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/14/2021] [Indexed: 12/20/2022]
Abstract
The COVID-19 (coronavirus disease) global pandemic, caused by the spread of the SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) virus, currently has limited treatment options which include vaccines, anti-virals, and repurposed therapeutics. With their high specificity, tunability, and biocompatibility, small molecules like peptides are positioned to act as key players in combating SARS-CoV-2, and can be readily modified to match viral mutation rate. A recent expansion of the understanding of the viral structure and entry mechanisms has led to the proliferation of therapeutic viral entry inhibitors. In this comprehensive review, inhibitors of SARS and SARS-CoV-2 are investigated and discussed based on therapeutic design, inhibitory mechanistic approaches, and common targets. Peptide therapeutics are highlighted, which have demonstrated in vitro or in vivo efficacy, discuss advantages of peptide therapeutics, and common strategies in identifying targets for viral inhibition.
Collapse
Affiliation(s)
- Disha Panchal
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Jeena Kataria
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Kamiya Patel
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Kaytlyn Crowe
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Varun Pai
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Abdul‐Rahman Azizogli
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Neil Kadian
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Sreya Sanyal
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Abhishek Roy
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Joseph Dodd‐o
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | | | - Vivek A. Kumar
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
- Department of Biomedical EngineeringDepartment of ChemicalBiological and Pharmaceutical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| |
Collapse
|