1
|
Rybarczyk A, Sultan T, Hussain N, Azam HMH, Rafique S, Zdarta J, Jesionowski T. Fusion of enzymatic proteins: Enhancing biological activities and facilitating biological modifications. Adv Colloid Interface Sci 2025; 340:103473. [PMID: 40086016 DOI: 10.1016/j.cis.2025.103473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
The fusion of enzymatic proteins represents a dynamic frontier in biotechnology and enzymatic engineering. This in-depth review looks at the many different ways that fusion proteins can be used, showing their importance in biosensing, gene therapy, targeted drug delivery, and biocatalysis. Fusion proteins have shown an astounding ability to improve and fine-tune biological functions by combining the most beneficial parts of different enzymes. Our first step is to explain how protein fusion increases biological functions. This will provide a broad picture of how this phenomenon has changed many fields. We dissect the intricate mechanisms through which fusion proteins orchestrate cellular processes, underscoring their potential to revolutionize the landscape of molecular biology. We also explore the complicated world of structural analysis and design strategies, stressing the importance of molecular insights for making the fusion protein approach work better. These insights broaden understanding of the underlying principles and illuminate the path toward unlocking untapped potential. The review also introduces cutting-edge techniques for constructing fusion protein libraries, such as DNA shuffling and phage display. These new methods allow scientists to build a molecular orchestra with an unprecedented level of accuracy, and thus use fusion proteins to their full potential in various situations. In conclusion, we provide a glimpse into the current challenges and prospects in fusion protein research, shedding light on recent advancements that promise to reshape the future of biotechnology. As we make this interesting journey through the field of enzymatic protein combination, it becomes clear that the fusion paradigm is about to start a new era of innovation that will push the limits of what is possible in biology and molecular engineering.
Collapse
Affiliation(s)
- Agnieszka Rybarczyk
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland
| | - Talha Sultan
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Nazim Hussain
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Hafiz Muhammad Husnain Azam
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Universitätsplatz 1, 01968 Senftenberg, Germany
| | - Safa Rafique
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Jakub Zdarta
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland.
| | - Teofil Jesionowski
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland.
| |
Collapse
|
2
|
Munusamy MA, Bharathi M, Alarfaj AA, Hussein-Al-Ali SH, Nagaiya R, Subbarayan S. Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles (GNANPs) induce antimicrobial properties and apoptosis in gastric cancer cells. Toxicol In Vitro 2025; 106:106037. [PMID: 40037499 DOI: 10.1016/j.tiv.2025.106037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 02/08/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
An assessment of the anticancer activity of Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles (GNANPs) against gastric cancer cells was the purpose of this study. The increasing prevalence of gastric cancer and the limitations of conventional therapies necessitate novel approaches that combine targeted drug delivery with therapeutic efficacy. Several techniques were used to characterize the synthesized GNANPs, including UV-visible spectroscopy, X-ray diffractometer (XRD), scanning electron microscope (SEM), transmission electron microscope (TEM), Fourier transform infrared (FT-IR), dynamic light scattering (DLS), and photoluminescence (PL). They were evaluated for their antimicrobial properties, cytotoxicity, ROS accumulation, apoptotic activity, and oxidative stress markers against AGS cells. The characterization analyses indicated the existence of Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles with an oval-shaped morphology and an average particle size of 127.80 nm. The existence of several elements and functional groups in the GNANPs was also detected using EDX and FT-IR analyses, respectively. The synthesized GNANPs have shown exceptional antibacterial activities by effectively inhibiting the growth of several infections. The treatment of GNANPs efficiently inhibited the growth of AGS cells. Fluorescence staining studies showed increased apoptosis and oxidative stress markers in AGS cells treated with synthesized Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles, indicating their potential as a viable cancer treatment option.
Collapse
Affiliation(s)
- Murugan Alwarkurichi Munusamy
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 602 105, India
| | - Muruganantham Bharathi
- Centre for Bioinformatics, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore-641021
| | - Abdullah A Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box.2455, Riyadh 11451, Saudi Arabia
| | | | - Ravichandran Nagaiya
- Department of Life Science and Environmental Biochemistry, Pusan National University, Busan 46241, Republic of Korea
| | - Sarathbabu Subbarayan
- Muthayammal Centre for Advanced Research, Muthayammal College of Arts and Science, Rasipuram, Namakkal, Tamil Nadu 637408, India.
| |
Collapse
|
3
|
Murphy G, Brayden DJ, Cheung DL, Liew A, Fitzgerald M, Pandit A. Albumin-based delivery systems: Recent advances, challenges, and opportunities. J Control Release 2025; 380:375-395. [PMID: 39842723 DOI: 10.1016/j.jconrel.2025.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/22/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Albumin and albumin-based biomaterials have been explored for various applications, including therapeutic delivery, as therapeutic agents, as components of tissue adhesives, and in tissue engineering applications. Albumin has been approved as a nanoparticle containing paclitaxel (Abraxane®), as an albumin-binding peptide (Victoza®), and as a glutaraldehyde-crosslinked tissue adhesive (BioGlue®). Albumin is also approved as a supportive therapy for various conditions, including hypoalbuminemia, sepsis, and acute respiratory distress syndrome (ARDS). However, no other new albumin-based systems in a hydrogel format have been used in the clinic. A review of publicly available clinical trials indicates that no new albumin drug delivery formats are currently in the clinical development pipeline. Although albumin has shown promise as a carrier of therapeutics for various diseases, including diabetes, cancers, and infectious diseases, its potential for treating blood-borne diseases such as HIV and leukemia has not been translated. This review offers a perspective on the use of albumin-based drug delivery systems for a broader range of disease applications, considering the protein properties and a review of the currently approved albumin-based technologies. This review supports ongoing efforts to advance biomedical research and clinical interventions through albumin-based delivery systems.
Collapse
Affiliation(s)
- Gillian Murphy
- CÚRAM, the Research Ireland Centre for Medical Devices, University of Galway, Ireland.
| | - David J Brayden
- CÚRAM, the Research Ireland Centre for Medical Devices, University of Galway, Ireland; School of Veterinary Medicine and Conway Institute, University College Dublin, Ireland
| | - David L Cheung
- CÚRAM, the Research Ireland Centre for Medical Devices, University of Galway, Ireland; School of Biological and Chemical Science, University of Galway, Ireland
| | - Aaron Liew
- Diabetes, Endocrinology and General Internal Medicine, Galway University Hospital, Galway, Ireland
| | | | - Abhay Pandit
- CÚRAM, the Research Ireland Centre for Medical Devices, University of Galway, Ireland.
| |
Collapse
|
4
|
Wang K, Sun M, Liu S, Wang R, Liu H, Qian F. Albumin-conjugated flumethasone for targeting and normalization of pancreatic stellate cells. J Control Release 2025; 380:994-1004. [PMID: 39983925 DOI: 10.1016/j.jconrel.2025.02.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/16/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
The tumor microenvironment (TME) plays a critical role in the poor clinical outlook for pancreatic ductal adenocarcinoma (PDAC). Activated pancreatic stellate cells (PSC) drive the complex interactions within the TME, resulting in a microenvironment that is resistant to chemotherapy and tolerant to the immune system, thereby promoting tumor growth. Effective deactivation of PSC is vital in treating pancreatic cancer. However, previous studies have only focused on limited changes in PSC phenotype without comprehensively analysing their overall function. Our transcriptome analysis identified agents capable of modulating multiple biological functions of PSC, including fibrosis, extracellular matrix generation, and the secretion of cytokines and immune factors. Through this comprehensive assessment, we discovered that flumethasone (Flu) effectively deactivates PSC. This glucocorticoid analogue remodels the tumor microenvironment by regulating the secretomes of PSC and their interaction with tumor cells. Additionally, our research revealed that activated PSC exhibited heightened albumin endocytosis. As a result, we propose that albumin conjugation could serve as an effective targeted drug delivery approach for PSC. Our findings also demonstrate that albumin-conjugated Flu maintained reprogramming capabilities in stromal cells, and enhanced the efficacy of chemotherapy in orthotopic mouse models of PDAC and KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre (KPC) pancreatic tumor allograft mouse model. Our investigation into the mechanism of PSC deactivation by flumethasone has revealed its potential for clinical cancer treatment through its effects on the tumor microenvironment. Furthermore, the conjugation of flumethasone to albumin enhances its safety and targeted delivery, offering a promising approach for PSC-targeted drug application in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Kaixin Wang
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Mengnan Sun
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Shiyu Liu
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Rui Wang
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Huiqin Liu
- Quaerite Biopharm Research, Beijing, China
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
5
|
Pongprayoon P, Kuntip N, Suwanasopee T, Jattawa D, Niramitranon J, Japrung D, Koonawootrittriron S. Comparative studies of structure and dynamics of caprine, leporine, ovine, and equine serum albumins. J Biomol Struct Dyn 2025; 43:2772-2780. [PMID: 38116752 DOI: 10.1080/07391102.2023.2294378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Serum albumin (SA) is the most prevalent protein found in blood. Human albumin was used as an albumin substitute in hypoalbuminemia pets due to high sequence similarity. SAs from furry animals were also reported to be the major indoor allergens. Sensitizing to one of SAs coupled with high sequence identity can lead to cross-reactive antibodies in allergic individuals. Thus, understanding the structural and dynamic characters of SAs is crucial for not only albumin substitution but also allergen therapy. Herein, Molecular dynamics (MD) simulations were performed to elucidate the structural and dynamic dissimilarity and similarity of economic animals [equine (ESA), caprine (CASA), ovine (OSA), and leporine (LSA)] to albumins from human (HSA), bovine (BSA), porcine (PSA), and pets [cat (FSA) and dog (CSA)]. The aim is to explore the feasibility of HSA substitution and understand how albumins cause the cross-reactivity. Generally, all albumins studied here show the scissoring motion like other mammalian albumins. The uniqueness of each albumin is defined by different sequence identity of domain I. Also, the drug binding affinity of studied albumins differs from HSA, CSA, FSA, BSA, and PSA. Especially, LSA displays the most deviated behavior from the group. So, such albumin may not be suitable for albumin therapy for pets and humans. CASA, OSA, and ESA share similar characteristics, therefore it is possible to use them to monitor the osmotic pressure among their species, but the allergenic response must be seriously considered. An insight obtained here can be useful to develop albumin therapy and understand clinical allergy.
Collapse
Affiliation(s)
- Prapasiri Pongprayoon
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
- Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| | - Nattapon Kuntip
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Thanathip Suwanasopee
- Department of Animal Science, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand
| | - Danai Jattawa
- Department of Animal Science, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand
| | - Jitti Niramitranon
- Department of Computer Engineering, Faculty of Engineering, Kasetsart University, Bangkok, Thailand
| | - Deanpen Japrung
- National Nanotechnology Center, National Science and Technology Development Agency, Pathumthani, Thailand
| | | |
Collapse
|
6
|
Shastri D, Raorane CJ, Raj V, Lee S. Human serum albumin-3-amino-1-propanesulfonic acid conjugate inhibits amyloid-β aggregation and mitigates cognitive decline in Alzheimer's disease. J Control Release 2025; 379:390-408. [PMID: 39805463 DOI: 10.1016/j.jconrel.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Alzheimer's disease (AD) is the most commonly occurring brain disorder, characterized by the accumulation of amyloid-β (Aβ) and tau, subsequently leading to neurocognitive decline. 3-Amino-1-propanesulfonic acid (TPS) and its prodrug, currently under clinical trial III, serve as promising therapeutic agents targeting Aβ pathology by specifically preventing monomer-to-oligomer formation. Inspired by the potency of TPS prodrug, we hypothesized that conjugating TPS with human serum albumin (HSA) could enhance brain delivery and synergistically inhibit Aβ aggregation in mild to moderate AD. Thus, we prepared and extensively characterized HSA-TPS (h-TPS) conjugate using an eco-friendly coupling method. In vitro studies on Aβ aggregation kinetics and AFM imaging revealed significant prevention of Aβ aggregation. Additionally, h-TPS significantly reduced Aβ-induced neurotoxicity and H2O2-mediated reactive oxygen species (ROS) stress in SH-SY5Y cells. Moreover, h-TPS administration improved blood-brain barrier permeability and cellular uptake into neuronal cells as well as showed in vivo uptake inside the brain within 1 h. In vivo studies using an Aβ1-42-induced acute AD rat model exhibited a dose-dependent significant reduction in hippocampal Aβ levels and restoration of declined spatial learning and memory with h-TPS treatment. Overall, findings suggest that h-TPS conjugate might be a promising neuroprotective agent for preventing Aβ aggregation in mild to moderate AD.
Collapse
Affiliation(s)
- Divya Shastri
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea; College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea
| | | | - Vinit Raj
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea.
| | - Sangkil Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Gulyaev IA, Sokol MB, Mollaeva MR, Klimenko MA, Yabbarov NG, Chirkina MV, Nikolskaya ED. Polymeric Drug Delivery Systems in Biomedicine. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S233-S262. [PMID: 40164161 DOI: 10.1134/s0006297924603976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 04/02/2025]
Abstract
Our review examines the key aspects of using polymeric carriers in biomedicine. The section "Polymers for Biomedicine" provides an overview of different types of polymers, their structural features and properties that determine their use as drug delivery vehicles. The section "Polymeric Carriers" characterizes the role of polymeric delivery systems in modern medicine. The main forms of polymeric carriers are described, as well as their key advantages for drug delivery. The section "Preclinical and Clinical Trials of Polymeric Drug Carriers" reviews the examples of clinical and preclinical studies of polymeric forms used for antitumor therapy, therapy for bacterial and infectious diseases. The final section "Targeted Drug Delivery Systems" is devoted to the discussion of approaches, as well as ligands that provide targeted drug delivery using polymeric carriers. We have paid special attention to modern approaches for increasing the efficacy of antibacterial therapy using vector molecules.
Collapse
Affiliation(s)
- Ivan A Gulyaev
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Maria B Sokol
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Mariia R Mollaeva
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Maksim A Klimenko
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Nikita G Yabbarov
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Margarita V Chirkina
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Elena D Nikolskaya
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia.
| |
Collapse
|
8
|
Wanselius M, Abrahmsén-Alami S, Hanafy BI, Mazza M, Hansson P. A microfluidic in vitro method predicting the fate of peptide drugs after subcutaneous administration. Int J Pharm 2024; 667:124849. [PMID: 39454976 DOI: 10.1016/j.ijpharm.2024.124849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
For many biopharmaceuticals, subcutaneous (sc) administration is the only viable route. However, there is no in vitro method available accurately predicting the absorption profiles of subcutaneously injected pharmaceuticals. In this work, we show that a recently developed microfluidics method for interaction studies (MIS) has the potential to be useful in this respect. The method utilises the responsiveness of polyelectrolyte microgel networks to oppositely charged molecules as a means to monitor the interaction between peptides and hyaluronic acid (HA), a major constituent of the subcutaneous extracellular matrix. We use the method to determine parameters describing the strength of interaction between peptide and HA as well as the peptide's aggregation tendency and transport properties in HA networks. The results from MIS studies of the peptide drugs exenatide, pramlintide, vancomycin, polymyxin B, lanreotide, MEDI7219 and AZD2820 are compared with results from measurements with the commercially available SCISSOR system and in vivo absorption and bioavailability data from the literature. We show that both MIS and SCISSOR reveal differences in the peptides' diffusivity and tendency to aggregate in the presence of HA. We show that MIS is particularly good at discriminating between peptides forming aggregates stabilised by non-electrostatic forces in the presence of HA, and peptides forming complexes stabilised by electrostatic interactions with HA. The method provides two parameters that can be used to quantify the peptides' aggregation tendency, the one describing the peptide packing density in complexes with HA and the other the apparent diffusivity upon release in a medium of physiological ionic strength and pH. The order of the peptides when ranked by increasing binding strength at pH 7.4 determined with MIS is shown to be in agreement with the order when ranked by the apparent 1st order absorption rate constant (ka) after sc administration in humans: lanreotide (Autogel) < exenatide (IRF) < AZD2820 < pramlintide < lanreotide (IRF) (IRF: Immediate release formulation). A correlation is found between the 1st order release rate constant determined with SCISSOR and ka for lanreotide (Autogel), exenatide and AZD2820. A mechanism relating the magnitude of ka to the peptides' charge is proposed.
Collapse
Affiliation(s)
- Marcus Wanselius
- Department of Medicinal Chemistry, Uppsala University, BMC P.O. Box 574, SE-751 23, Uppsala, Sweden
| | - Susanna Abrahmsén-Alami
- Innovation Strategy & External Liaison. Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Belal I Hanafy
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge, United Kingdom
| | - Mariarosa Mazza
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge, United Kingdom
| | - Per Hansson
- Department of Medicinal Chemistry, Uppsala University, BMC P.O. Box 574, SE-751 23, Uppsala, Sweden.
| |
Collapse
|
9
|
Fernández-Sainz J, Herrera-Ochoa D, Pacheco-Liñán PJ, Darder M, Albaladejo J, Bravo I, Garzón-Ruiz A. Spectroscopic study on volasertib: Highly stable complexes with albumin and encapsulation into alginate/montmorillonite bionanocomposites. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 322:124823. [PMID: 39033609 DOI: 10.1016/j.saa.2024.124823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/05/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
In the present work, we study different physicochemical properties related to LADME processes of volasertib, a Polo-like kinase 1 inhibitor in advanced clinical trials. Firstly, the protonation equilibria, the extent of ionization at the physiological pH and pKa values of this drug are studied combining spectroscopic techniques and computational calculations. Secondly, the binding process of volasertib to the human serum albumin (HSA) protein is analyzed by fluorescence spectroscopy. We report a high binding constant to HSA (Ka = 4.10 × 106 M-1) and their pharmacokinetic implications are discussed accordingly. The negative enthalpy and entropy (ΔH0 = -54.49 kJ/mol; ΔS0 = -58.90 J K-1 mol-1) determined for the binding process suggests the implication of hydrogen bonds and van der Waals interactions in the formation of the HSA-volasertib complex. Additionally, volasertib is encapsulated in an alginate/montmorillonite bionanocomposite as a proof of concept for an oral delivery nanocarrier. The physical properties of that nanocomposite as well as volasertib delivery kinetics are analyzed.
Collapse
Affiliation(s)
- Jesús Fernández-Sainz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Diego Herrera-Ochoa
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Pedro J Pacheco-Liñán
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Margarita Darder
- Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Cantoblanco, 28049 Madrid, Spain
| | - José Albaladejo
- Departamento de Química Física, Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain
| | - Iván Bravo
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Andrés Garzón-Ruiz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain.
| |
Collapse
|
10
|
Gholijani N, Azarpira N, Abolmaali SS, Tanideh N, Ravanrooy MH, Taki F, Daryabor G. Piperine and piperine-loaded albumin nanoparticles ameliorate adjuvant-induced arthritis and reduce IL-17 in rats. Exp Mol Pathol 2024; 140:104937. [PMID: 39353355 DOI: 10.1016/j.yexmp.2024.104937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/03/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
AIM Rheumatoid arthritis (RA) is one of the most common chronic, inflammatory, autoimmune diseases affecting mainly the joints. Piperine (PIP), an alkaloid found in black pepper, has anti-inflammatory properties and its use in drug delivery systems such as nanoparticles might be a treatment for RA. This study aims to evaluate the possible anti-inflammatory and anti-arthritic effects of PIP and its use in albumin nanoparticles as a possible approach for the treatment of Adjuvant-induced arthritis (AIA) rats. METHODS PIP-loaded Bovine Serum Albumin nanoparticles (PIP-BSA NPs) were prepared using a desolvation method. AIA rats were given intraperitoneal injections of either 40 mg PIP or 131 mg PIP-BSA NPs every two days until day 28 when animals were sacrificed. Clinical score, histopathology, X-ray radiography, and serum levels of pro-inflammatory cytokines such as IL-1β, IL-17, and TNF-α were evaluated. RESULTS PIP and PIP-BSA NPs significantly reduced clinical scores, and alleviated inflammation within the joints. PIP was superior to PIP-BSA NPs for the alleviation of fibrin deposition and periosteal reactions while bone inflammation and erosion were less severe in the case of PIP-BSA NPs. Besides, both of the treatments suppressed serum levels of IL-17 in AIA rats (p = 0.003 and p = 0.02; respectively). CONCLUSIONS PIP and PIP-BSA NPs effectively alleviate the severity of AIA and suppress inflammation. Due to the superiority of PIP in improving fibrin deposition and periosteal reactions and the efficacy of PIP-BSA NPs in suppressing bone inflammation and erosion, their simultaneous use might be investigated.
Collapse
Affiliation(s)
- Nasser Gholijani
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira-Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology and Nanotechnology in Drug Delivery Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Tanideh
- Stem Cells Technology Research Center, Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Farzane Taki
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Daryabor
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
11
|
Han J, Shen Y, Cao R, Wang W, Duan J, Duan J, Bao C. Active herbal ingredients and drug delivery design for tumor therapy: a review. Chin J Nat Med 2024; 22:1134-1162. [PMID: 39725513 DOI: 10.1016/s1875-5364(24)60741-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Indexed: 12/28/2024]
Abstract
Active herbal ingredients are gaining recognition for their potent anti-tumor efficacy, attributable to various mechanisms including tumor cell inhibition, immune system activation, and tumor angiogenesis inhibition. Recent studies have revealed that numerous anti-tumor herbal ingredients, such as ginsenosides, ursolic acid, oleanolic acid, and Angelica sinensis polysaccharides, can be utilized to develop smart drug carriers like liposomes, micelles, and nanoparticles. These carriers can deliver active herbal ingredients and co-deliver anti-tumor drugs to enhance drug accumulation at tumor sites, thereby improving anti-tumor efficacy. This study provides a comprehensive analysis of the mechanisms by which these active herbal ingredients-derived carriers enhance therapeutic outcomes. Additionally, it highlights the structural properties of these active herbal ingredients, demonstrating how their unique features can be strategically employed to design smart drug carriers with improved anti-tumor efficacy. The insights presented aim to serve as a reference and guide future innovations in the design and application of smart drug carriers for cancer therapy that leverage active herbal ingredients.
Collapse
Affiliation(s)
- Jing Han
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yanxi Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruiying Cao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiren Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jialun Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Chunjie Bao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
12
|
Cho YJ, Kim H, Lim SI. Preserved structure and function of human serum albumin self-folded in the oxidative cytoplasm of Escherichia coli. J Biotechnol 2024; 390:62-70. [PMID: 38761885 DOI: 10.1016/j.jbiotec.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
Human serum albumin (HSA), a polypeptide featuring 17 disulfide bonds, acts as a crucial transport protein in human blood plasma. Its extended circulation half-life, mediated by FcRn (neonatal Fc receptor)-facilitated recycling, positions HSA as an excellent carrier for long-acting drug delivery. However, the conventional method of obtaining HSA from human blood faces limitations due to availability and potential contamination risks, such as blood-borne diseases. This study introduced SHuffle, an oxidative Escherichia coli (E. coli) expression system, for the production of recombinant HSA (rHSA) that spontaneously self-folds into its native conformation. This system ensures precise disulfide bond formation and correct folding of cysteine-rich rHSA, eliminating the need for chaperone co-expression or domain fusion of a folding enhancer. The purified rHSA underwent thorough physicochemical characterization, including mass spectrometry, circular dichroism spectroscopy, intrinsic fluorescence spectroscopy, esterase-like activity assay, and size exclusion chromatography, to assess critical quality attributes. Importantly, rHSA maintained native binding affinity to FcRn and the albumin-binding domain. Collectively, our analyses demonstrated a high comparability between rHSA and plasma-derived HSA. The expression of rHSA in E. coli with an oxidizing cytosol provides a secure and cost-effective approach, enhancing the potential of rHSA for diverse medical applications.
Collapse
Affiliation(s)
- Yong Joon Cho
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Busan, Republic of Korea
| | - Hyunji Kim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Busan, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Busan, Republic of Korea.
| |
Collapse
|
13
|
Wei Q, Wu Y, Jiang X, Lu W, Liu S, Yu J. Supramolecular prodrug of SN38 based on endogenous albumin and SN38 prodrug modified with semaglutide side chain to improve the tumor distribution. Bioorg Med Chem 2024; 106:117754. [PMID: 38728869 DOI: 10.1016/j.bmc.2024.117754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
To improve the biodistribution of the drug in the tumor, a supramolecular prodrug of SN38 was fabricated in situ between endogenous albumin and SN38 prodrug modified with semaglutide side chain. Firstly, SN38 was conjugated with semaglutide side chain and octadecanedioic acid via glycine linkers to obtain SI-Gly-SN38 and OA-Gly-SN38 prodrugs, respectively. Both SI-Gly-SN38 and OA-Gly-SN38 exhibited excellent stability in PBS for over 24 h. Due to the strong binding affinity of the semaglutide side chain with albumin, the plasma half-life of SI-Gly-SN38 was 2.7 times higher than that of OA-Gly-SN38. Furthermore, with addition of HSA, the fluorescence intensity of SI-Gly-SN38 was 4 times higher than that of OA-Gly-SN38, confirming its strong binding capability with HSA. MTT assay showed that the cytotoxicity of SI-Gly-SN38 and OA-Gly-SN38 was higher than that of Irinotecan. Even incubated with HSA, the SI-Gly-SN38 and OA-Gly-SN38 still maintained high cytotoxicity, indicating minimal influence of HSA on their cytotoxicity. In vivo pharmacokinetic studies demonstrated that the circulation half-life of SI-Gly-SN38 was twice that of OA-Gly-SN38. SI-Gly-SN38 exhibited significantly reduced accumulation in the lungs, being only 0.23 times that of OA-Gly-SN38. The release of free SN38 in the lungs from SI-Gly-SN38 was only 0.4 times that from OA-Gly-SN38 and Irinotecan. The SI-Gly-SN38 showed the highest accumulation in tumors. The tumor inhibition rate of SI-Gly-SN38 was 6.42% higher than that of OA-Gly-SN38, and 8.67% higher than that of Irinotecan, respectively. These results indicate that the supramolecular prodrug delivery system can be constructed between SI-Gly-SN38 and endogenous albumin, which improves drug biodistribution in vivo, enhances tumor accumulation, and plays a crucial role in tumor growth inhibition.
Collapse
Affiliation(s)
- Qingyu Wei
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yanyan Wu
- Department of Radiology, the Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xing Jiang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Shiyuan Liu
- Department of Radiology, the Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| | - Jiahui Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
14
|
Berger O, Choi W, Ko CH, Thompson MP, Avram MJ, Scott DJ, Hoare BL, Cridge R, Wheatley M, Bathgate RAD, Batlle D, Gianneschi NC. Long-Circulating Vasoactive 1,18-Octadecanedioic Acid-Terlipressin Conjugate. ACS Pharmacol Transl Sci 2024; 7:1252-1261. [PMID: 38751631 PMCID: PMC11092119 DOI: 10.1021/acsptsci.3c00305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/29/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024]
Abstract
Hepatorenal syndrome (HRS) is a life-threatening complication of end-stage liver disease first reported over a century ago, but its management still poses an unmet challenge. A therapeutic agent found to stabilize the condition is a short cyclic peptide, vasopressin analogue, terlipressin (TP). While TP is commonly prescribed for HRS patients in most parts of the world, it was only recently approved for use in the United States. TP exhibits short circulation half-lives and adverse side effects associated with the dose required. Herein, we present a 1,18-octadecanedioic acid (ODDA) conjugate of the cyclic peptide (ODDA-TP), which enables noncovalent binding to serum albumin via native fatty acid binding modes. ODDA-TP is demonstrated to outperform TP alone in studies including in vitro cellular receptor activation, stability in plasma, pharmacokinetics, and performance in vivo in rats. Specifically, ODDA-TP had an elimination half-life 20 times that of TP alone while exhibiting a superior safety profile.
Collapse
Affiliation(s)
- Or Berger
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Wonmin Choi
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Caroline H. Ko
- NewCures,
Innovation and Ventures Office, Northwestern
University, Evanston, Illinois 60208, United States
| | - Matthew P. Thompson
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Michael J. Avram
- Feinberg
Medical School, Northwestern University, Chicago, Illinois 60611, United States
- Department
of Anesthesiology, Northwestern University, Chicago, Illinois 60611, United States
| | - Daniel J. Scott
- The
Florey,Parkville, Victoria 3010, Australia
- Department
of Biochemistry and Pharmacology, The University
of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | - Mark Wheatley
- Centre
for Sport, Exercise and Life Sciences, Coventry
University, Coventry CV1 5FB, U.K.
- Centre
of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands B15 2TT, U.K.
| | - Ross A. D. Bathgate
- The
Florey,Parkville, Victoria 3010, Australia
- Department
of Biochemistry and Pharmacology, The University
of Melbourne, Parkville, Victoria 3010, Australia
| | - Daniel Batlle
- Feinberg
Medical School, Northwestern University, Chicago, Illinois 60611, United States
- Department
of Medicine Division of Nephrology and Hypertension, Chicago, Illinois 60611, United States
| | - Nathan C. Gianneschi
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department
of Materials Science and Engineering, Northwestern
University, Evanston, Illinois 60208, United States
- Department of Biomedical Engineering, Northwestern
University, Evanston, Illinois 60208, United States
- Department of Pharmacology, Northwestern
University, Chicago, Illinois 60611, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Simpson-Querrey Institute, Northwestern
University, Chicago, Illinois 60611, United States
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
15
|
Kuna K, Baddam SR, Kalagara S, Akkiraju PC, Tade RS, Enaganti S. Emerging natural polymer-based architectured nanotherapeutics for the treatment of cancer. Int J Biol Macromol 2024; 262:129434. [PMID: 38232877 DOI: 10.1016/j.ijbiomac.2024.129434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/20/2023] [Accepted: 01/10/2024] [Indexed: 01/19/2024]
Abstract
The field of cancer therapy is advancing rapidly, placing a crucial emphasis on innovative drug delivery systems. The increasing global impact of cancer highlights the need for creative therapeutic strategies. Natural polymer-based nanotherapeutics have emerged as a captivating avenue in this pursuit, drawing substantial attention due to their inherent attributes. These attributes include biodegradability, biocompatibility, negligible toxicity, extended circulation time, and a wide range of therapeutic payloads. The unique size, shape, and morphological characteristics of these systems facilitate profound tissue penetration, complementing active and passive targeting strategies. Moreover, these nanotherapeutics exploit specific cellular and subcellular trafficking pathways, providing precise control over drug release kinetics. This comprehensive review emphasizes the utilization of naturally occurring polymers such as polysaccharides (e.g., chitosan, hyaluronic acid, alginates, dextran, and cyclodextrin) and protein-based polymers (e.g., ferritin, gelatin, albumin) as the foundation for nanoparticle development. The paper meticulously examines their in vitro characteristics alongside in vivo efficacy, particularly focusing on their pivotal role in ameliorating diverse types of solid tumors within cancer therapy. The amalgamation of material science ingenuity and biological insight has led to the formulation of these nanoparticles, showcasing their potential to reshape the landscape of cancer treatment.
Collapse
Affiliation(s)
- Krishna Kuna
- Department of Chemistry, University College of Science, Saifabad, Osmania University, Hyderabad, Telangana, India.
| | - Sudhakar Reddy Baddam
- University of Massachusetts Chan Medical School, RNA Therapeutics Institute, Worcester, MA 01655, United States of America
| | - Sudhakar Kalagara
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave, El Paso, TX 79968, United States of America
| | - Pavan C Akkiraju
- Department of Biotechnology, School of Allied Healthcare Sciences, Malla Reddy University, Hyderabad, India
| | - Rahul S Tade
- Department of Pharmaceutics, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405, India
| | - Sreenivas Enaganti
- Department of Bioinformatics, Averinbiotech Laboratories, Nallakunta, Hyderabad, Telangana, India
| |
Collapse
|
16
|
Kopp A, Kwon H, Johnston C, Vance S, Legg J, Galson-Holt L, Thurber GM. Impact of tissue penetration and albumin binding on design of T cell targeted bispecific agents. Neoplasia 2024; 48:100962. [PMID: 38183712 PMCID: PMC10809211 DOI: 10.1016/j.neo.2023.100962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024]
Abstract
Bispecific agents are a rapidly growing class of cancer therapeutics, and immune targeted bispecific agents have the potential to expand functionality well beyond monoclonal antibody agents. Humabodies⁎ are fully human single domain antibodies that can be linked in a modular fashion to form multispecific therapeutics. However, the effect of heterogeneous delivery on the efficacy of crosslinking bispecific agents is currently unclear. In this work, we utilize a PSMA-CD137 Humabody with an albumin binding half-life extension (HLE) domain to determine the impact of tissue penetration on T cell activating bispecific agents. Using heterotypic spheroids, we demonstrate that increased tissue penetration results in higher T cell activation at sub-saturating concentrations. Next, we tested the effect of two different albumin binding moieties on tissue distribution using albumin-specific HLE domains with varying affinities for albumin and a non-specific lipophilic dye. The results show that a specific binding mechanism to albumin does not influence tissue penetration, but a non-specific mechanism reduced both spheroid uptake and distribution in the presence of albumin. These results highlight the potential importance of tissue penetration on bispecific agent efficacy and describe how the design parameters including albumin-binding domains can be selected to maximize the efficacy of bispecific agents.
Collapse
Affiliation(s)
- Anna Kopp
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Hyeyoung Kwon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | | | | | - James Legg
- Crescendo Biologics, Cambridge, United Kingdom
| | | | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, United States
| |
Collapse
|
17
|
Nagar N, Naidu G, Mishra A, Poluri KM. Protein-Based Nanocarriers and Nanotherapeutics for Infection and Inflammation. J Pharmacol Exp Ther 2024; 388:91-109. [PMID: 37699711 DOI: 10.1124/jpet.123.001673] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023] Open
Abstract
Infectious and inflammatory diseases are one of the leading causes of death globally. The status quo has become more prominent with the onset of the coronavirus disease 2019 (COVID-19) pandemic. To combat these potential crises, proteins have been proven as highly efficacious drugs, drug targets, and biomarkers. On the other hand, advancements in nanotechnology have aided efficient and sustained drug delivery due to their nano-dimension-acquired advantages. Combining both strategies together, the protein nanoplatforms are equipped with the advantageous intrinsic properties of proteins as well as nanoformulations, eloquently changing the field of nanomedicine. Proteins can act as carriers, therapeutics, diagnostics, and theranostics in their nanoform as fusion proteins or as composites with other organic/inorganic materials. Protein-based nanoplatforms have been extensively explored to target the major infectious and inflammatory diseases of clinical concern. The current review comprehensively deliberated proteins as nanocarriers for drugs and nanotherapeutics for inflammatory and infectious agents, with special emphasis on cancer and viral diseases. A plethora of proteins from diverse organisms have aided in the synthesis of protein-based nanoformulations. The current study specifically presented the proteins of human and pathogenic origin to dwell upon the field of protein nanotechnology, emphasizing their pharmacological advantages. Further, the successful clinical translation and current bottlenecks of the protein-based nanoformulations associated with the infection-inflammation paradigm have also been discussed comprehensively. SIGNIFICANCE STATEMENT: This review discusses the plethora of promising protein-based nanocarriers and nanotherapeutics explored for infectious and inflammatory ailments, with particular emphasis on protein nanoparticles of human and pathogenic origin with reference to the advantages, ADME (absorption, distribution, metabolism, and excretion parameters), and current bottlenecks in development of protein-based nanotherapeutic interventions.
Collapse
Affiliation(s)
- Nupur Nagar
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Goutami Naidu
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Amit Mishra
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| |
Collapse
|
18
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
19
|
Ashraf S, Qaiser H, Tariq S, Khalid A, Makeen HA, Alhazmi HA, Ul-Haq Z. Unraveling the versatility of human serum albumin - A comprehensive review of its biological significance and therapeutic potential. Curr Res Struct Biol 2023; 6:100114. [PMID: 38111902 PMCID: PMC10726258 DOI: 10.1016/j.crstbi.2023.100114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/20/2023] Open
Abstract
Human serum albumin (HSA) is a multi-domain macromolecule with diverse ligand binding capability because of its ability to allow allosteric modulation despite being a monomeric protein. Physiologically, HSA act as the primary carrier for various exogenous and endogenous compounds and fatty acids, and alter the pharmacokinetic properties of several drugs. It has antioxidant properties and is utilized therapeutically to improve the drug delivery of pharmacological agents for the treatment of several disorders. The flexibility of albumin in holding various types of drugs coupled with a variety of modifications makes this protein a versatile drug carrier with incalculable potential in therapeutics. This review provides a brief outline of the different structural properties of HSA, and its various binding sites, moreover, an overview of the genetic, biomedical, and allosteric modulation of drugs and drug delivery aspects of HSA is also included, which may be helpful in guiding advanced clinical applications and further research on the therapeutic potential of this extraordinary protein.
Collapse
Affiliation(s)
- Sajda Ashraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75210, Pakistan
| | - Hina Qaiser
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75210, Pakistan
| | - Sumayya Tariq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75210, Pakistan
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan, Saudi Arabia
- Medicinal and Aromatic Plants Research Institute, National Center for Research, P.O. Box: 2424, Khartoum, 11111, Sudan
| | - Hafiz A. Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A. Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75210, Pakistan
| |
Collapse
|
20
|
McCarthy DR, Remington JM, Ferrell JB, Schneebeli ST, Li J. Nano-Bio Interactions between DNA Nanocages and Human Serum Albumin. J Chem Theory Comput 2023; 19:7873-7881. [PMID: 37877553 PMCID: PMC11070245 DOI: 10.1021/acs.jctc.3c00720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
DNA nanostructures have emerged as promising nanomedical tools due to their biocompatibility and tunable behavior. Recent work has shown that DNA nanocages decorated with organic dendrimers strongly bind human serum albumin (HSA), yet the dynamic structures of these complexes remain uncharacterized. This theoretical and computational investigation elucidates the fuzzy interactions between dendritically functionalized cubic DNA nanocages and HSA. The dendrimer-HSA interactions occur via nonspecific binding with the protein thermodynamically and kinetically free to cross the open faces of the cubic scaffold. However, the rigidity of the DNA scaffold prevents the binding energetics from scaling with the number of dendrimers. These discoveries not only provide a useful framework by which to model general interactions of DNA nanostructures complexed with serum proteins but also give valuable molecular insight into the design of next-generation DNA nanomedicines.
Collapse
Affiliation(s)
| | | | | | - Severin T. Schneebeli
- Department of Chemistry, University of Vermont, Burlington, VT 05405
- Department of Industrial and Physical Pharmacy and Department of Chemistry, Purdue University, West Lafayette, IN 47907
| | - Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT 05405
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907
| |
Collapse
|
21
|
Garg U, Jain N, Kaul S, Nagaich U. Role of Albumin as a Targeted Drug Carrier in the Management of Rheumatoid Arthritis: A Comprehensive Review. Mol Pharm 2023; 20:5345-5358. [PMID: 37870420 DOI: 10.1021/acs.molpharmaceut.3c00581] [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: 10/24/2023]
Abstract
An endogenous transporter protein called albumin interacts with the Fc receptor to provide it with multiple substrate-binding domains, cell membrane receptor activation, and an extended circulating half-life. Albumin has the remarkable ability to bind with receptors viz. secreted protein acidic and rich in cysteine (SPARC) and scavenger protein-A (SR-A) that are overexpressed during rheumatoid arthritis (RA), enabling active targeting of the disease site instead of requiring specialized substrates to be added to the nanocarrier. RA, a chronic autoimmune illness, is characterized by the presence of a severe inflammatory response. RA patients have low serum albumin concentration, which signifies the high uptake of albumin at the inflammatory sites, giving a rationale to use albumin as a drug carrier for RA therapy. Albumin has the capacity for both passive and active targeting. It is an abundantly available protein in the bloodstream showing excellent cellular compatibility, degradability in biological tissues, nonantigenicity, and safety. There are three strategies of albumin mediated drug delivery as encapsulating therapeutics in albumin nanoparticles, chemically conjugating drugs with functional proteins, and albumin itself which is used as a targeting ligand to deliver drugs specifically to cells or tissues that express albumin-binding receptors. In the current review, an attempt has been made to highlight the significant evidence of albumin as a drug delivery carrier for the safe and effective management of RA. Evidence has been provided in the form of recent research advances, clinical trials, and patents. Additionally, this review will outline the prospective for the potential utilization of albumin as a drug vehicle for RA and suggest possible future avenues to provide the perspective for subsequent studies.
Collapse
Affiliation(s)
- Unnati Garg
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| | - Neha Jain
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| | - Shreya Kaul
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| | - Upendra Nagaich
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| |
Collapse
|
22
|
Xu C, Shen H, Liu TM, Kwok RT, Lam JW, Tang BZ. Restriction of molecular motion to a higher level: Towards bright AIE dots for biomedical applications. iScience 2023; 26:106568. [PMID: 37128609 PMCID: PMC10148129 DOI: 10.1016/j.isci.2023.106568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
In the late 19th century, scientists began to study the photophysical differences between chromophores in the solution and aggregate states, which breed the recognition of the prototypical processes of aggregation-caused quenching and aggregation-induced emission (AIE). In particular, the conceptual discovery of the AIE phenomenon has spawned the innovation of luminogenic materials with high emission in the aggregate state based on their unique working principle termed the restriction of intramolecular motion. As AIE luminogens have been practically fabricated into AIE dots for bioimaging, further improvement of their brightness is needed although this is technically challenging. In this review, we surveyed the recent advances in strategic molecular engineering of highly emissive AIE dots, including nanoscale crystallization and matrix-assisted rigidification. We hope that this timely summary can deepen the understanding about the root cause of the high emission of AIE dots and provide inspiration to the rational design of functional aggregates.
Collapse
Affiliation(s)
- Changhuo Xu
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional Materials, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Hanchen Shen
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional Materials, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao, China
| | - Ryan T.K. Kwok
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional Materials, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jacky W.Y. Lam
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, Guangdong-Hong Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional Materials, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Key Laboratory of Functional Aggregate, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
- Center for Aggregation-Induced Emission, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
23
|
Liang Z, Sun Y, Zeng H, Qin H, Yang R, Qu L, Zhang K, Li Z. Broad-Specificity Screening of Pyrethroids Enabled by the Catalytic Function of Human Serum Albumin on Coumarin Hydrolysis. Anal Chem 2023; 95:5678-5686. [PMID: 36952638 DOI: 10.1021/acs.analchem.2c05556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
Sensing systems based on cholinesterase and carboxylesterase coupled with different transduction technologies have emerged for pesticide screening owing to their simple operation, fast response, and suitability for on-site analysis. However, the broad spectrum and specificity screening of pyrethroids over organophosphates and carbamates remains an unmet challenge for current enzymatic sensors. Human serum albumin (HSA), a multifunctional protein, can promote various chemical transformations and show a high affinity for pyrethroids, which offer a route for specific and broad-spectrum pyrethroid screening. Herein, for the first time, we evaluated the catalytic hydrolysis function of human serum albumin (HSA) on the coumarin lactone bond and revealed that HSA can act as an enzyme to catalyze the hydrolysis of the coumarin lactone bond. Molecular docking and chemical modifications indicate that lysine 199 and tyrosine 411 serve as the catalytic general base and contribute to most of the catalytic activity. Utilizing this enzymatic activity, a broad specific ratiometric fluorescence pyrethroids sensing system was developed. The binding energetics and binding constants of pesticides and HSA show that pyrethroids bind to HSA more easily than organophosphates and carbamates, which is responsible for the specificity of the sensing system. This study provides a general sensor platform and strategy for screening pesticides and reveals the catalytic activity of HSA on the hydrolysis of the coumarin lactone bond, which may open innovative horizons for the chemical sensing and biomedical applications of HSA.
Collapse
Affiliation(s)
- Zengqiang Liang
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanqiang Sun
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Huajin Zeng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Haimei Qin
- Fujian Provincial Key Lab of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Ran Yang
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Lingbo Qu
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Food Safety Quick Testing and Smart Supervision Technology for State Market Regulation, Zhengzhou 450001, China
| | - Ke Zhang
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Zhaohui Li
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
24
|
Fernández-Sainz J, Pacheco-Liñán PJ, Ripoll C, González-Fuentes J, Albaladejo J, Bravo I, Garzón-Ruiz A. Unusually High Affinity of the PLK Inhibitors RO3280 and GSK461364 to HSA and Its Possible Pharmacokinetic Implications. Mol Pharm 2023; 20:1631-1642. [PMID: 36812406 PMCID: PMC9997069 DOI: 10.1021/acs.molpharmaceut.2c00849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The binding processes of two Polo-like kinase inhibitors, RO3280 and GSK461364, to the human serum albumin (HSA) protein as well as the protonation equilibria of both compounds have been studied combining absorbance and fluorescence spectroscopy experiments together with density functional theory calculations. We found that the charge states of RO3280 and GSK461364 are +2 and +1, respectively, at the physiological pH. Nevertheless, RO3280 binds to HSA in the charge state +1 prior to a deprotonation pre-equilibrium. Binding constants to site I of HSA of 2.23 × 106 and 8.80 × 104 M-1 were determined for RO3280 and GSK461364, respectively, at 310 K. The binding processes of RO3280 and GSK461364 to HSA are entropy- and enthalpy-driven, respectively. The positive enthalpy found for the RO3280-HSA complex formation could be related to a proton pre-equilibrium of RO3280.
Collapse
Affiliation(s)
- Jesús Fernández-Sainz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Pedro J Pacheco-Liñán
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Consuelo Ripoll
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Joaquín González-Fuentes
- Centro Regional de Investigaciones Biomédicas (CRIB), Unidad Asociada de Biomedicina (UCLM-CSIC), C/ Almansa, 14, 02008 Albacete, Spain
| | - José Albaladejo
- Departamento de Química Física, Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain
| | - Iván Bravo
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain.,Centro Regional de Investigaciones Biomédicas (CRIB), Unidad Asociada de Biomedicina (UCLM-CSIC), C/ Almansa, 14, 02008 Albacete, Spain
| | - Andrés Garzón-Ruiz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| |
Collapse
|
25
|
Logesh K, Raj B, Bhaskaran M, Thirumaleshwar S, Gangadharappa H, Osmani R, Asha Spandana K. Nanoparticulate drug delivery systems for the treatment of rheumatoid arthritis: A comprehensive review. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
26
|
RB Singh K, Nagpure G, Singh J, Singh RP. Introduction to drug-delivery techniques based on nanotechnological approaches. NANOTECHNOLOGY FOR DRUG DELIVERY AND PHARMACEUTICALS 2023:3-28. [DOI: 10.1016/b978-0-323-95325-2.00010-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
27
|
Mali G, Maji S, Chavan KA, Shukla M, Kumar M, Bhattacharyya S, Erande RD. Effective Synthesis and Biological Evaluation of Functionalized 2,3-Dihydrofuro[3,2- c]coumarins via an Imidazole-Catalyzed Green Multicomponent Approach. ACS OMEGA 2022; 7:36028-36036. [PMID: 36249391 PMCID: PMC9558716 DOI: 10.1021/acsomega.2c05361] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/16/2022] [Indexed: 06/01/2023]
Abstract
For the first time, an eco-friendly and efficient one-pot green multicomponent approach has been described to synthesize functionalized trans-2,3-dihydrofuro[3,2-c]coumarins (DHFCs). In this synthesis, imidazole and water were used as the catalyst and solvent, respectively, under mild conditions. Applications of the developed catalytic process in a water medium revealed the outstanding activity, productivity, and broad functional group tolerance, affording a series of newly designed DHFC and derivatives in excellent yields (72-98%). Moreover, the human serum albumin (HSA) binding ability of the synthesized DHFC derivatives has been uncovered through the detailed in silico and in vitro-based structure-activity analysis. The ability to bind HSA, the most abundant serum protein, in the low micromolar ranges unequivocally reflects the suitable absorption, distribution, metabolism, and elimination profile of the synthesized compounds, which may further be envisaged for their therapeutic usage endeavors.
Collapse
Affiliation(s)
- Ghanshyam Mali
- Department
of Chemistry, Indian Institute of Technology
Jodhpur, Jodhpur 342037, India
| | - Sushobhan Maji
- Department
of Bioscience and Bioengineering, Indian
Institute of Technology Jodhpur, Jodhpur 342037, India
| | - Kailas Arjun Chavan
- Department
of Chemistry, Indian Institute of Technology
Jodhpur, Jodhpur 342037, India
| | - Manjari Shukla
- Department
of Bioscience and Bioengineering, Indian
Institute of Technology Jodhpur, Jodhpur 342037, India
| | - Manish Kumar
- Department
of Chemistry, Indian Institute of Technology
Jodhpur, Jodhpur 342037, India
| | - Sudipta Bhattacharyya
- Department
of Bioscience and Bioengineering, Indian
Institute of Technology Jodhpur, Jodhpur 342037, India
| | - Rohan D. Erande
- Department
of Chemistry, Indian Institute of Technology
Jodhpur, Jodhpur 342037, India
| |
Collapse
|
28
|
Ullah A, Kwon HT, Lim SI. Albumin: A Multi-talented Clinical and Pharmaceutical Player. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-022-0104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Baldari S, Di Modugno F, Nisticò P, Toietta G. Strategies for Efficient Targeting of Tumor Collagen for Cancer Therapy. Cancers (Basel) 2022; 14:cancers14194706. [PMID: 36230627 PMCID: PMC9563908 DOI: 10.3390/cancers14194706] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary The tumor microenvironment encompasses the cellular and extracellular matrix components that support and shape the three-dimensional framework in which solid tumors develop and grow. The extracellular matrix of the tumor is characterized by increased deposition and aberrant architecture of collagen fibers. Therefore, as a key mechanical component of the tumor microenvironment, collagen plays a critical role in cancer progression, metastasis, and therapeutic response. To boost the efficacy of current anticancer therapies, including immunotherapy, innovative approaches should take into account strategies directed against the dysregulated non-cancer cell stromal components. In the current review, we provide an overview of the principal approaches to target tumor collagen to provide therapeutic benefits. Abstract The tumor stroma, which comprises stromal cells and non-cellular elements, is a critical component of the tumor microenvironment (TME). The dynamic interactions between the tumor cells and the stroma may promote tumor progression and metastasis and dictate resistance to established cancer therapies. Therefore, novel antitumor approaches should combine anticancer and anti-stroma strategies targeting dysregulated tumor extracellular matrix (ECM). ECM remodeling is a hallmark of solid tumors, leading to extensive biochemical and biomechanical changes, affecting cell signaling and tumor tissue three-dimensional architecture. Increased deposition of fibrillar collagen is the most distinctive alteration of the tumor ECM. Consequently, several anticancer therapeutic strategies have been developed to reduce excessive tumor collagen deposition. Herein, we provide an overview of the current advances and challenges of the main approaches aiming at tumor collagen normalization, which include targeted anticancer drug delivery, promotion of degradation, modulation of structure and biosynthesis of collagen, and targeting cancer-associated fibroblasts, which are the major extracellular matrix producers.
Collapse
|
30
|
Varga N, Seres L, Kovács NA, Turcsányi Á, Juhász Á, Csapó E. Serum albumin/hyaluronic acid nanoconjugate: Evaluation of concentration-dependent structural changes to form an efficient drug carrier particle. Int J Biol Macromol 2022; 220:1523-1531. [PMID: 36122775 DOI: 10.1016/j.ijbiomac.2022.09.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Norbert Varga
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - László Seres
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - Nikolett Alexandra Kovács
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - Árpád Turcsányi
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - Ádám Juhász
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - Edit Csapó
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary.
| |
Collapse
|
31
|
Computation-Aided Design of Albumin Affibody-Inserted Antibody Fragment for the Prolonged Serum Half-Life. Pharmaceutics 2022; 14:pharmaceutics14091769. [PMID: 36145517 PMCID: PMC9500697 DOI: 10.3390/pharmaceutics14091769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Single-chain variable fragments (scFvs) have been recognized as promising agents in cancer therapy. However, short serum half-life of scFvs often limits clinical application. Fusion to albumin affibody (ABD) is an effective and convenient half-life extension strategy. Although one terminus of scFv is available for fusion of ABD, it is also frequently used for fusion of useful moieties such as small functional proteins, cytokines, or antibodies. Herein, we investigated the internal linker region for ABD fusion instead of terminal region, which was rarely explored before. We constructed two internally ABD-inserted anti-HER2 4D5scFv (4D5-ABD) variants, which have short (4D5-S-ABD) and long (4D5-L-ABD) linker length respectively. The model structures of these 4D5scFv and 4D5-ABD variants predicted using the deep learning-based protein structure prediction program (AlphaFold2) revealed high similarity to either the original 4D5scFv or the ABD structure, implying that the functionality would be retained. Designed 4D5-ABD variants were expressed in the bacterial expression system and characterized. Both 4D5-ABD variants showed anti-HER2 binding affinity comparable with 4D5scFv. Binding affinity of both 4D5-ABD variants against albumin was also comparable. In a pharmacokinetic study in mice, the 4D5-ABD variants showed a significantly prolonged half-life of 34 h, 114 times longer than that of 4D5scFv. In conclusion, we have developed a versatile scFv platform with enhanced pharmacokinetic profiles with an aid of deep learning-based structure prediction.
Collapse
|
32
|
Curcio M, Vittorio O, Bell JL, Iemma F, Nicoletta FP, Cirillo G. Hyaluronic Acid within Self-Assembling Nanoparticles: Endless Possibilities for Targeted Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12162851. [PMID: 36014715 PMCID: PMC9413373 DOI: 10.3390/nano12162851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 05/27/2023]
Abstract
Self-assembling nanoparticles (SANPs) based on hyaluronic acid (HA) represent unique tools in cancer therapy because they combine the HA targeting activity towards cancer cells with the advantageous features of the self-assembling nanosystems, i.e., chemical versatility and ease of preparation and scalability. This review describes the key outcomes arising from the combination of HA and SANPs, focusing on nanomaterials where HA and/or HA-derivatives are inserted within the self-assembling nanostructure. We elucidate the different HA derivatization strategies proposed for this scope, as well as the preparation methods used for the fabrication of the delivery device. After showing the biological results in the employed in vivo and in vitro models, we discussed the pros and cons of each nanosystem, opening a discussion on which approach represents the most promising strategy for further investigation and effective therapeutic protocol development.
Collapse
Affiliation(s)
- Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Orazio Vittorio
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Jessica Lilian Bell
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Francesca Iemma
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
33
|
Dou L, Liu H, Wang K, Liu J, Liu L, Ye J, Wang R, Deng H, Qian F. Albumin binding revitalizes NQO1 bioactivatable drugs as novel therapeutics for pancreatic cancer. J Control Release 2022; 349:876-889. [PMID: 35907592 DOI: 10.1016/j.jconrel.2022.07.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/10/2022] [Accepted: 07/24/2022] [Indexed: 11/16/2022]
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1) is an enzyme significantly overexpressed in pancreatic ductal adenocarcinoma (PDAC) tumors compared to the associated normal tissues. NQO1 bioactivatable drugs, such as β-lapachone (β-lap), can be catalyzed to generate reactive oxygen species (ROS) for direct tumor killing. However, the extremely narrow therapeutic window caused by methemoglobinemia and hemolytic anemia severely restricts its further clinical translation despite considerable efforts in the past 20 years. Previously, we demonstrated that albumin could be utilized to deliver cytotoxic drugs selectively into KRAS-mutant PDAC with a much expanded therapeutic window due to KRAS-enhanced macropinocytosis and reduced neonatal Fc receptor (FcRn) expression in PDAC. Herein, we analyzed the expression patterns of albumin and FcRn across major organs in LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre (KPC) mice. The tumors were the predominant tissues with both elevated albumin and reduced FcRn expression, thus making them an ideal target for albumin-based drug delivery. Quantitative proteomics analysis of tissue samples from 5 human PDAC patients further confirmed the elevated albumin/FcRn ratio. Given such a compelling biological rationale, we designed a nanoparticle albumin-bound prodrug of β-lap, nab-(pro-β-lap), to achieve PDAC targeted delivery and expand the therapeutic window of β-lap. We found that nab-(pro-β-lap) uptake was profoundly enhanced by KRAS mutation. Compared to the solution formulation of the parent drug β-lap, nab-(pro-β-lap) showed enhanced safety due to much lower rates of methemoglobinemia and hemolytic anemia, which was confirmed both in vitro and in vivo. Furthermore, nab-(pro-β-lap) significantly inhibited tumor growth in subcutaneously implanted KPC xenografts and enhanced the pharmacodynamic endpoints (e.g., PARP1 hyperactivation, γ-H2AX). Thus, nab-(pro-β-lap), with improved safety and antitumor efficacy, offers a drug delivery strategy with translational viability for β-lap in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Lei Dou
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Huiqin Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Kaixin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Jing Liu
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, PR China
| | - Lei Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Junxiao Ye
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Rui Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, PR China
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
34
|
Fab fragment immobilized immunoaffinity cryogels as a tool for human serum albumin purification: Characterization of Fab immobilized cryogels. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1203:123311. [DOI: 10.1016/j.jchromb.2022.123311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
|
35
|
Plumet C, Châtre R, Djago F, Péraudeau E, Blancart-Remaury Q, Clarhaut J, Geffroy C, Said Mohamed A, Opalinski I, Renoux B, Poinot P, Papot S. A β-Cyclodextrin-Albumin Conjugate for Enhancing Therapeutic Efficacy of Cytotoxic Drugs. Bioconjug Chem 2022; 33:1138-1144. [PMID: 35613473 DOI: 10.1021/acs.bioconjchem.2c00182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Enhancing the selectivity of anticancer drugs currently used in the clinic is of great interest in order to propose more efficient chemotherapies with fewer side effects for patients. In this context, we developed a β-cyclodextrin trimer that binds to circulating albumin to form the corresponding bioconjugate in the bloodstream. This latter can then entrap doxorubicin following its i.v. administration via the formation of a host-guest inclusion complex and deliver the drug in tumors. In this study, we demonstrate that the β-cyclodextrin trimer improves the therapeutic efficacy of doxorubicin for the treatment of a subcutaneous murine Lewis lung carcinoma (LLC) implanted in C57BL/6 mice. This outcome is associated with an increased deposition of doxorubicin in malignant tissues when used in combination with the β-cyclodextrin trimer compared to the administration of the drug alone.
Collapse
Affiliation(s)
- Chad Plumet
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Rémi Châtre
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Fabiola Djago
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Elodie Péraudeau
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Quentin Blancart-Remaury
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Jonathan Clarhaut
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France.,CHU de Poitiers, 2 rue de la Miléterie, CS 90577, 86021 Poitiers, France
| | - Claude Geffroy
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Achmet Said Mohamed
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Isabelle Opalinski
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Brigitte Renoux
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Pauline Poinot
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France
| | - Sébastien Papot
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, cedex 9, France.,Seekyo SA, 2 avenue Galilée, BP 30153, 86961 Futuroscope, France
| |
Collapse
|
36
|
Cho H, Jeon SI, Ahn CH, Shim MK, Kim K. Emerging Albumin-Binding Anticancer Drugs for Tumor-Targeted Drug Delivery: Current Understandings and Clinical Translation. Pharmaceutics 2022; 14:728. [PMID: 35456562 PMCID: PMC9028280 DOI: 10.3390/pharmaceutics14040728] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/20/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Albumin has shown remarkable promise as a natural drug carrier by improving pharmacokinetic (PK) profiles of anticancer drugs for tumor-targeted delivery. The exogenous or endogenous albumin enhances the circulatory half-lives of anticancer drugs and passively target the tumors by the enhanced permeability and retention (EPR) effect. Thus, the albumin-based drug delivery leads to a potent antitumor efficacy in various preclinical models, and several candidates have been evaluated clinically. The most successful example is Abraxane, an exogenous human serum albumin (HSA)-bound paclitaxel formulation approved by the FDA and used to treat locally advanced or metastatic tumors. However, additional clinical translation of exogenous albumin formulations has not been approved to date because of their unexpectedly low delivery efficiency, which can increase the risk of systemic toxicity. To overcome these limitations, several prodrugs binding endogenous albumin covalently have been investigated owing to distinct advantages for a safe and more effective drug delivery. In this review, we give account of the different albumin-based drug delivery systems, from laboratory investigations to clinical applications, and their potential challenges, and the outlook for clinical translation is discussed. In addition, recent advances and progress of albumin-binding drugs to move more closely to the clinical settings are outlined.
Collapse
Affiliation(s)
- Hanhee Cho
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Korea
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Seong Ik Jeon
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Cheol-Hee Ahn
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Man Kyu Shim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Kwangmeyung Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|
37
|
Hatami E, Nagesh PKB, Chauhan N, Jaggi M, Chauhan SC, Yallapu MM. In Situ Nanoparticle Self-Assembly for Combination Delivery of Therapeutics to Non-Small Cell Lung Cancer. ACS APPLIED BIO MATERIALS 2022; 5:1104-1119. [PMID: 35179871 DOI: 10.1021/acsabm.1c01158] [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] [Indexed: 11/28/2022]
Abstract
Chemotherapy often experiences several challenges including severe systemic toxicity and adverse effects. The combination chemotherapy arose as an effective clinical practice aimed at reducing doses of drugs to achieve synergistic actions with low toxicity. Our recent efforts demonstrated a synergistic therapeutic benefit of gambogic acid (GA) and gemcitabine (Gem) against lung cancer. However, simultaneous delivery of these two drugs at the tumor site is highly challenging. Therefore, the development of an injectable formulation that can effectively deliver both hydrophobic (GA) and hydrophilic (Gem) drugs in one formulation is a clinically unmet need. Herein, this study reports an in situ human serum albumin (HSA)- and tannic acid (TA)-mediated complexed GA and Gem nanoparticles (G-G@HTA NPs). G-G@HTA NP formation was confirmed by the particle size, Fourier transform infrared spectroscopy, and 1H NMR spectroscopy. The superior therapeutic activity of G-G@HTA NPs was demonstrated by multiple in vitro functional assays. Additionally, G-G@HTA NPs revealed an obvious and precise targeting of tumors in vivo. The promoted and more synergistic anti-tumor efficacy of G-G@HTA NPs was attained than that of combined treatments and single drug treatments. These events have resulted in no apparent systemic and organ toxicities. Together, this study suggests that in situ HSA-TA-based combinatorial treatment strategy is a suitable approach for application in lung cancer treatment.
Collapse
Affiliation(s)
- Elham Hatami
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Neeraj Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| |
Collapse
|
38
|
Brandt F, Ullrich M, Laube M, Kopka K, Bachmann M, Löser R, Pietzsch J, Pietzsch HJ, van den Hoff J, Wodtke R. "Clickable" Albumin Binders for Modulating the Tumor Uptake of Targeted Radiopharmaceuticals. J Med Chem 2021; 65:710-733. [PMID: 34939412 DOI: 10.1021/acs.jmedchem.1c01791] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The intentional binding of radioligands to albumin gains increasing attention in the context of radiopharmaceutical cancer therapy as it can lead to an enhanced radioactivity uptake into the tumor lesions and, thus, to a potentially improved therapeutic outcome. However, the influence of the radioligand's albumin-binding affinity on the time profile of tumor uptake has been only partly addressed so far. Based on the previously identified Nε-4-(4-iodophenyl)butanoyl-lysine scaffold, we designed "clickable" lysine-derived albumin binders (cLABs) and determined their dissociation constants toward albumin by novel assay methods. Structure-activity relationships were derived, and selected cLABs were applied for the modification of the somatostatin receptor subtype 2 ligand (Tyr3)octreotate. These novel conjugates were radiolabeled with copper-64 and subjected to a detailed in vitro and in vivo radiopharmacological characterization. Overall, the results of this study provide an incentive for further investigations of albumin binders for applications in endoradionuclide therapies.
Collapse
Affiliation(s)
- Florian Brandt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Martin Ullrich
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Markus Laube
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
| | - Reik Löser
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Hans-Jürgen Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Jörg van den Hoff
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Technische Universität Dresden, Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
| | - Robert Wodtke
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| |
Collapse
|
39
|
Sharma S, Parveen R, Chatterji BP. Toxicology of Nanoparticles in Drug Delivery. CURRENT PATHOBIOLOGY REPORTS 2021; 9:133-144. [PMID: 34840918 PMCID: PMC8611175 DOI: 10.1007/s40139-021-00227-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/19/2021] [Indexed: 12/17/2022]
Abstract
Nanoparticles have revolutionized biomedicine especially in the field of drug delivery due to their intriguing properties such as systemic stability, level of solubility, and target site specificity. It can, however, be both beneficial and damaging depending on the properties in different environments, thus highlighting the importance of nanotoxicology studies before use in humans. Different types of nanoparticles have been used in drug delivery, and this review summarizes the recent toxicity studies of these nanoparticles. The toxicological evaluation of three widely used nanoparticles in drug delivery that are metal, lipid, and protein nanoparticles has been discussed in detail. Studies have recorded several toxic effects of various nanoparticles such as metal-based nanoparticles have been linked to increased oxidative stress and have the potential to infiltrate the cell nucleus and protein-based nanoparticles have been observed to have hepatotoxicity and nephrotoxicity as their adverse effects. Considering the increasing application of nanoparticles in drug delivery and the growing concerns of regulatory authorities regarding the toxicity of nanocarriers in living organisms, it requires urgent attention to identify the gap in toxicity studies. The review highlights the gap in toxicity studies and potential focus areas to overcome the existing challenges.
Collapse
Affiliation(s)
- Swati Sharma
- St. Xavier's College, Mumbai, Maharashtra 400001 India
| | - Roza Parveen
- School of Engineering, Ajeenkya DY Patil University, Pune, Maharashtra 412105 India
| | | |
Collapse
|
40
|
Yang B, Kwon I. Chemical Modification of Cysteine with 3-Arylpropriolonitrile Improves the In Vivo Stability of Albumin-Conjugated Urate Oxidase Therapeutic Protein. Biomedicines 2021; 9:biomedicines9101334. [PMID: 34680451 PMCID: PMC8533278 DOI: 10.3390/biomedicines9101334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
3-arylpropiolonitriles (APN) are promising alternatives to maleimide for chemo-selective thiol conjugation, because the reaction product has a remarkably hydrolytic stability compared with that of thiol-maleimide reactions in vitro. However, whether cysteine modification with APN enhances stability in vivo compared to thiol-maleimide reactions remains unclear, probably due to the too short in vivo serum half-life of a protein to observe significant cleavage of thiol-maleimide/-APN reaction products. The conjugation of human serum albumin (HSA) to a therapeutic protein reportedly prolongs the in vivo serum half-life. To evaluate the in vivo stability of the thiol-APN reaction product, we prepared HSA-conjugated Arthrobacter globiformis urate oxidase (AgUox), a therapeutic protein for gout treatment. Site-specific HSA conjugation to AgUox was achieved by combining site-specific incorporation of tetrazine containing an amino acid (frTet) into AgUox and a crosslinker containing trans-cyclooctene and either thiol-maleimide (AgUox-MAL-HSA) or -APN chemistry (AgUox-APN-HSA). Substantial cleavage of the thioester of AgUox-MAL-HSA was observed in vitro, whereas no cleavage of the thiol-APN product of AgUox-APN-HSA was observed. Furthermore, the in vivo serum half-life of AgUox-APN-HSA in the late phase was significantly longer than that of AgUox-MAL-HSA. Overall, these results demonstrate that the thiol-APN chemistry enhanced the in vivo stability of the HSA-conjugated therapeutic protein.
Collapse
|
41
|
Lechner VM, Nappi M, Deneny PJ, Folliet S, Chu JCK, Gaunt MJ. Visible-Light-Mediated Modification and Manipulation of Biomacromolecules. Chem Rev 2021; 122:1752-1829. [PMID: 34546740 DOI: 10.1021/acs.chemrev.1c00357] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chemically modified biomacromolecules-i.e., proteins, nucleic acids, glycans, and lipids-have become crucial tools in chemical biology. They are extensively used not only to elucidate cellular processes but also in industrial applications, particularly in the context of biopharmaceuticals. In order to enable maximum scope for optimization, it is pivotal to have a diverse array of biomacromolecule modification methods at one's disposal. Chemistry has driven many significant advances in this area, and especially recently, numerous novel visible-light-induced photochemical approaches have emerged. In these reactions, light serves as an external source of energy, enabling access to highly reactive intermediates under exceedingly mild conditions and with exquisite spatiotemporal control. While UV-induced transformations on biomacromolecules date back decades, visible light has the unmistakable advantage of being considerably more biocompatible, and a spectrum of visible-light-driven methods is now available, chiefly for proteins and nucleic acids. This review will discuss modifications of native functional groups (FGs), including functionalization, labeling, and cross-linking techniques as well as the utility of oxidative degradation mediated by photochemically generated reactive oxygen species. Furthermore, transformations at non-native, bioorthogonal FGs on biomacromolecules will be addressed, including photoclick chemistry and DNA-encoded library synthesis as well as methods that allow manipulation of the activity of a biomacromolecule.
Collapse
Affiliation(s)
- Vivian M Lechner
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Manuel Nappi
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Patrick J Deneny
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Sarah Folliet
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - John C K Chu
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Matthew J Gaunt
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
42
|
Modelling and Control of Corticotropin Permeation from Hydrogels across a Natural Membrane in the Presence of Albumin. Processes (Basel) 2021. [DOI: 10.3390/pr9091674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
(1) Background: Skin is a difficult barrier to overcome, especially for molecules with masses greater than 500 Da. It has been suggested that albumin may contribute to more effective penetration of many therapeutic substances. In this study, an attempt was made to use albumin in semi-solid formulations to increase the skin penetration of another peptide—corticotropin (ACTH). (2) Methods: Hydrogels were prepared at two concentrations: 15 mg/g and 20 mg/g corticotropin, then albumin was added to them in different stoichiometric ratios. The degree of ACTH release from hydrogels, both with and without albumin addition, was investigated. For selected hydrogels the process of corticotropin permeation through a model membrane, i.e., pig skin, was examined. (3) Results: The study of corticotropin release showed that the addition of albumin, depending on its amount, may delay or increase the release process. Similarly, a study of ACTH permeation through porcine skin showed that albumin can delay or increase and accelerate ACTH permeation. (4) Conclusions: Hydrogel, applicated on the skin surface, may prove to be a beneficial and convenient solution for patients. It is an innovative way of application ACTH that bypasses the gastrointestinal tract and may result in increased availability of the peptide and its efficacy.
Collapse
|
43
|
Thermostable and Long-Circulating Albumin-Conjugated Arthrobacter globiformis Urate Oxidase. Pharmaceutics 2021; 13:pharmaceutics13081298. [PMID: 34452259 PMCID: PMC8400835 DOI: 10.3390/pharmaceutics13081298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Urate oxidase derived from Aspergillus flavus has been investigated as a treatment for tumor lysis syndrome, hyperuricemia, and gout. However, its long-term use is limited owing to potential immunogenicity, low thermostability, and short circulation time in vivo. Recently, urate oxidase isolated from Arthrobacter globiformis (AgUox) has been reported to be thermostable and less immunogenic than the Aspergillus-derived urate oxidase. Conjugation of human serum albumin (HSA) to therapeutic proteins has become a promising strategy to prolong circulation time in vivo. To develop a thermostable and long-circulating urate oxidase, we investigated the site-specific conjugation of HSA to AgUox based on site-specific incorporation of a clickable non-natural amino acid (frTet) and an inverse electron demand Diels-Alder reaction. We selected 14 sites for frTet incorporation using the ROSETTA design, a computational stability prediction program, among which AgUox containing frTet at position 196 (Ag12) exhibited enzymatic activity and thermostability comparable to those of wild-type AgUox. Furthermore, Ag12 exhibited a high HSA conjugation yield without compromising the enzymatic activity, generating well-defined HSA-conjugated AgUox (Ag12-HSA). In mice, the serum half-life of Ag12-HSA was approximately 29 h, which was roughly 17-fold longer than that of wild-type AgUox. Altogether, this novel formulated AgUox may hold enhanced therapeutic efficacy for several diseases.
Collapse
|
44
|
Kudłacik-Kramarczyk S, Drabczyk A, Głąb M, Gajda P, Czopek A, Zagórska A, Jaromin A, Gubernator J, Makara A, Tyliszczak B. The Development of the Innovative Synthesis Methodology of Albumin Nanoparticles Supported by Their Physicochemical, Cytotoxic and Hemolytic Evaluation. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4386. [PMID: 34442909 PMCID: PMC8400698 DOI: 10.3390/ma14164386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 01/28/2023]
Abstract
Many studies are being performed to develop effective carriers for controlled cytostatic delivery wherein albumin is a promising material due to its tendency to accumulate near cancer cells. The novelty of this work involves the development of the synthesis methodology of albumin nanoparticles and their biological and physicochemical evaluation. Albumin particles were obtained via the salt-induced precipitation and K3PO4 was used as a salting-out agent. Various concentrations of protein and salting-out agent solutions were mixed using a burette or a syringe system. It was proved that the size of the particles depended on the concentrations of the reagents and the methodology applied. As a result of a process performed using a burette and 2 M K3PO4, albumin spheres having a size 5-25 nm were obtained. The size of nanospheres and their spherical shape was confirmed via TEM analysis. The use of a syringe system led to preparation of particles of large polydispersity. The highest albumin concentration allowing for synthesis of homogeneous particles was 2 g/L. The presence of albumin in spheres was confirmed via the FT-IR technique and UV-Vis spectroscopy. All samples showed no cytotoxicity towards normal human dermal fibroblasts and no hemolytic properties against human erythrocytes (the hemolysis did not exceed 2.5%).
Collapse
Affiliation(s)
- Sonia Kudłacik-Kramarczyk
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Anna Drabczyk
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Magdalena Głąb
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Paweł Gajda
- Department of Sustainable Energy Development, Faculty of Energy and Fuels, AGH University of Science and Technology, 30 Mickiewicza Av., 30-059 Krakow, Poland;
| | - Anna Czopek
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (A.C.); (A.Z.)
| | - Agnieszka Zagórska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (A.C.); (A.Z.)
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 14a Joliot-Curie St., 50-383 Wroclaw, Poland; (A.J.); (J.G.)
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 14a Joliot-Curie St., 50-383 Wroclaw, Poland; (A.J.); (J.G.)
| | - Agnieszka Makara
- Faculty of Chemical Engineering and Technology, Cracow University of Technology, 24 Warszawska St., 31-155 Krakow, Poland;
| | - Bożena Tyliszczak
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| |
Collapse
|
45
|
Song M, Fu W, Liu Y, Yao H, Zheng K, Liu L, Xue J, Xu P, Chen Y, Huang M, Li J. Unveiling the molecular mechanism of pH-dependent interactions of human serum albumin with chemotherapeutic agent doxorubicin: A combined spectroscopic and constant-pH molecular dynamics study. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.115949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Turtoi M, Anghelache M, Patrascu AA, Maxim C, Manduteanu I, Calin M, Popescu DL. Synthesis, Characterization, and In Vitro Insulin-Mimetic Activity Evaluation of Valine Schiff Base Coordination Compounds of Oxidovanadium(V). Biomedicines 2021; 9:562. [PMID: 34067862 PMCID: PMC8156053 DOI: 10.3390/biomedicines9050562] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 11/17/2022] Open
Abstract
Type 2 diabetes became an alarming global health issue since the existing drugs do not prevent its progression. Herein, we aimed to synthesize and characterize a family of oxidovanadium(V) complexes with Schiff base ligands derived from L-/D-valine (val) and salicylaldehyde (sal) or o-vanillin (van) as insulin-mimetic agents and to assess their potential anti-diabetic properties. Two new oxidovanadium(V) complexes, [{VVO(R-salval)(H2O)}(μ2-O){VVO(R-salval)}] and [{VVO(R-vanval)(CH3OH)}2(μ2-O)], and their S-enantiomers were synthesized and characterized. The compounds exhibit optical activity as shown by crystallographic and spectroscopic data. The stability, the capacity to bind bovine serum albumin (BSA), the cytotoxicity against human hepatoma cell line, as well as the potential anti-diabetic activity of the four compounds are investigated. The synthesized compounds are stable for up to three hours in physiological conditions and exhibit a high capacity of binding to BSA. Furthermore, the synthesized compounds display cytocompatibility at biologically relevant concentrations, exert anti-diabetic potential and insulin-mimetic activities by inhibiting the α-amylase and protein tyrosine phosphatase activity, and a long-term increase of insulin receptor phosphorylation compared to the insulin hormone. Thus, the in vitro anti-diabetic potential and insulin-mimetic properties of the newly synthesized oxidovanadium(V) compounds, correlated with their cytocompatibility, make them promising candidates for further investigation as anti-diabetic drugs.
Collapse
Affiliation(s)
- Mihaela Turtoi
- Medical and Pharmaceutical Bionanotechnologies Laboratory, Institute of Cellular Biology and Pathology Nicolae Simionescu of the Romanian Academy, 8 B.P. Hasdeu, 050568-Bucharest, Romania; (M.A.); (I.M.)
| | - Maria Anghelache
- Medical and Pharmaceutical Bionanotechnologies Laboratory, Institute of Cellular Biology and Pathology Nicolae Simionescu of the Romanian Academy, 8 B.P. Hasdeu, 050568-Bucharest, Romania; (M.A.); (I.M.)
| | - Andrei A. Patrascu
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Bucharest, 23 Dumbrava Roşie, 020464-Bucharest, Romania; (A.A.P.); (C.M.)
| | - Catalin Maxim
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Bucharest, 23 Dumbrava Roşie, 020464-Bucharest, Romania; (A.A.P.); (C.M.)
| | - Ileana Manduteanu
- Medical and Pharmaceutical Bionanotechnologies Laboratory, Institute of Cellular Biology and Pathology Nicolae Simionescu of the Romanian Academy, 8 B.P. Hasdeu, 050568-Bucharest, Romania; (M.A.); (I.M.)
| | - Manuela Calin
- Medical and Pharmaceutical Bionanotechnologies Laboratory, Institute of Cellular Biology and Pathology Nicolae Simionescu of the Romanian Academy, 8 B.P. Hasdeu, 050568-Bucharest, Romania; (M.A.); (I.M.)
| | - Delia-Laura Popescu
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Bucharest, 23 Dumbrava Roşie, 020464-Bucharest, Romania; (A.A.P.); (C.M.)
| |
Collapse
|
47
|
Hashimoto K. CD137 as an Attractive T Cell Co-Stimulatory Target in the TNFRSF for Immuno-Oncology Drug Development. Cancers (Basel) 2021; 13:2288. [PMID: 34064598 PMCID: PMC8150789 DOI: 10.3390/cancers13102288] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/24/2022] Open
Abstract
Immune checkpoint inhibitors have altered the treatment landscape significantly in several cancers, yet not enough for many cancer patients. T cell costimulatory receptors have been pursued as targets for the next generation of cancer immunotherapies, however, sufficient clinical efficacy has not yet been achieved. CD137 (TNFRSF9, 4-1BB) provides co-stimulatory signals and activates cytotoxic effects of CD8+ T cells and helps to form memory T cells. In addition, CD137 signalling can activate NK cells and dendritic cells which further supports cytotoxic T cell activation. An agonistic monoclonal antibody to CD137, urelumab, provided promising clinical efficacy signals but the responses were achieved above the maximum tolerated dose. Utomilumab is another CD137 monoclonal antibody to CD137 but is not as potent as urelumab. Recent advances in antibody engineering technologies have enabled mitigation of the hepato-toxicity that hampered clinical application of urelumab and have enabled to maintain similar potency to urelumab. Next generation CD137 targeting molecules currently in clinical trials support T cell and NK cell expansion in patient samples. CD137 targeting molecules in combination with checkpoint inhibitors or ADCC-enhancing monoclonal antibodies have been sought to improve both clinical safety and efficacy. Further investigation on patient samples will be required to provide insights to understand compensating pathways for future combination strategies involving CD137 targeting agents to optimize and maintain the T cell activation status in tumors.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Crescendo Biologics, Ltd., Meditrina Building 260, Babraham Research Campus, Cambridge CB22 3AT, UK
| |
Collapse
|
48
|
Abstract
Since the last decade, the polymer-drug conjugate (PDC) approach has emerged as one of the most promising drug-delivery technologies owing to several benefits like circumventing premature drug release, offering controlled and targeted drug delivery, improving the stability, safety, and kinetics of conjugated drugs, and so forth. In recent years, PDC technology has advanced with the objective to further enhance the treatment outcomes by integrating nanotechnology and multifunctional characteristics into these systems. One such development is the ability of PDCs to act as theranostic agents, permitting simultaneous diagnosis and treatment options. Theranostic nanocarriers offer the opportunity to track the distribution of PDCs within the body and help to localize the diseased site. This characteristic is of particular interest, especially among those therapeutic approaches where external stimuli are supposed to be applied for abrupt drug release at the target site for localized delivery to avoid systemic side effects (e.g., Visudyne®). Thus, with the help of this review article, we are presenting the most recent updates in the domain of PDCs as nanotheranostic agents. Different methodologies utilized to design PDCs along with imaging characteristics and their applicability in a wide range of diseases, have been summarized in this article.
Collapse
|