151
|
Han J, Pan C, Tang X, Li Q, Zhu Y, Zhang Y, Liang A. Hypersensitivity reactions to small molecule drugs. Front Immunol 2022; 13:1016730. [PMID: 36439170 PMCID: PMC9684170 DOI: 10.3389/fimmu.2022.1016730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/20/2022] [Indexed: 02/02/2024] Open
Abstract
Drug hypersensitivity reactions induced by small molecule drugs encompass a broad spectrum of adverse drug reactions with heterogeneous clinical presentations and mechanisms. These reactions are classified into allergic drug hypersensitivity reactions and non-allergic drug hypersensitivity reactions. At present, the hapten theory, pharmacological interaction with immune receptors (p-i) concept, altered peptide repertoire model, and altered T-cell receptor (TCR) repertoire model have been proposed to explain how small molecule drugs or their metabolites induce allergic drug hypersensitivity reactions. Meanwhile, direct activation of mast cells, provoking the complement system, stimulating or inhibiting inflammatory reaction-related enzymes, accumulating bradykinin, and/or triggering vascular hyperpermeability are considered as the main factors causing non-allergic drug hypersensitivity reactions. To date, many investigations have been performed to explore the underlying mechanisms involved in drug hypersensitivity reactions and to search for predictive and preventive methods in both clinical and non-clinical trials. However, validated methods for predicting and diagnosing hypersensitivity reactions to small molecule drugs and deeper insight into the relevant underlying mechanisms are still limited.
Collapse
Affiliation(s)
- Jiayin Han
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chen Pan
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuan Tang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Zhu
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yushi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aihua Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
152
|
Protein corona mediated liposomal drug delivery for bacterial infection management. Asian J Pharm Sci 2022; 17:855-866. [PMID: 36600900 PMCID: PMC9800951 DOI: 10.1016/j.ajps.2022.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/10/2022] [Accepted: 10/15/2022] [Indexed: 11/07/2022] Open
Abstract
Liposomes have been widely investigated as a class of promising antibiotic delivery systems for the treatment of life-threatening bacterial infections. However, the inevitable formation of protein corona on the liposomal surface can heavily impact in vivo performance. A better understanding of the effects of protein corona on liposomal behavior can significantly improve antibacterial liposomal drug development. Here, the critical role of protein corona in mediating liposome-bacteria interactions was elucidated. Adsorption of negatively charged protein on cationic liposome weakened electrostatic attraction-enhanced liposomal binding to the bacteria. Cumulative complement deposition on anionic liposome composed of phosphatidylglycerol (DSPG sLip) contributed to a superior binding affinity of DSPG sLip to planktonic bacteria and biofilms, which was exploited to enhance bacteria-targeted drug delivery. In both S. aureus-related osteomyelitis and pneumonia mice models, DSPG sLip was demonstrated as a promising antibiotic nanocarrier for managing MRSA infection, indicating the benefits of lipid composition-based protein corona modulation in liposomal antibiotic delivery for bacterial infection treatment.
Collapse
|
153
|
Pourmadadi M, Abbasi P, Eshaghi MM, Bakhshi A, Ezra Manicum AL, Rahdar A, Pandey S, Jadoun S, Díez-Pascual AM. Curcumin delivery and co-delivery based on nanomaterials as an effective approach for cancer therapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
154
|
Pisani A, Donno R, Valenti G, Pompa PP, Tirelli N, Bardi G. Chemokine-Decorated Nanoparticles Target Specific Subpopulations of Primary Blood Mononuclear Leukocytes. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3560. [PMID: 36296750 PMCID: PMC9609899 DOI: 10.3390/nano12203560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
Specific cell targeting to deliver nanoparticles can be achieved by tailored modifications of the material surface with chemical moieties. The selection of the cell targets can be optimized by covering the nanoparticle with molecules, the receptor expression of which is restricted to particular cell subsets. Chemokines perform their biological action through 7-TM Gi-protein-coupled receptors differently expressed in all tissues. We decorated the surface of biocompatible polymer nanoparticles with full-length CCL5, an inflammatory chemokine that attracts leukocytes by binding CCR5, which is highly expressed in blood-circulating monocytes. Our observations showed that CCL5 functionalization does not affect the nanoparticle biocompatibility. Notably, CCL5 NPs delivered to PBMCs are selectively internalized by CCR5+ monocytes but not by CCR5- lymphocytes. The efficacy of PBMC subpopulation targeting by chemokine-decorated nanoparticles establishes an easy-to-use functionalization for specific leukocyte delivery.
Collapse
Affiliation(s)
- Anissa Pisani
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
- Department of Chemistry and Industrial Chemistry, University of Genova, Via Dodecaneso 31, 16146 Genova, Italy
| | - Roberto Donno
- Laboratory of Polymers and Biomaterials, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Giulio Valenti
- Laboratory of Polymers and Biomaterials, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Pier Paolo Pompa
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Nicola Tirelli
- Laboratory of Polymers and Biomaterials, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Giuseppe Bardi
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
155
|
Dobrovolskaia MA. Lessons learned from immunological characterization of nanomaterials at the Nanotechnology Characterization Laboratory. Front Immunol 2022; 13:984252. [PMID: 36304452 PMCID: PMC9592561 DOI: 10.3389/fimmu.2022.984252] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Nanotechnology carriers have become common in pharmaceutical products because of their benefits to drug delivery, including reduced toxicities and improved efficacy of active pharmaceutical ingredients due to targeted delivery, prolonged circulation time, and controlled payload release. While available examples of reduced drug toxicity through formulation using a nanocarrier are encouraging, current data also demonstrate that nanoparticles may change a drug’s biodistribution and alter its toxicity profile. Moreover, individual components of nanoparticles and excipients commonly used in formulations are often not immunologically inert and contribute to the overall immune responses to nanotechnology-formulated products. Said immune responses may be beneficial or adverse depending on the indication, dose, dose regimen, and route of administration. Therefore, comprehensive toxicology studies are of paramount importance even when previously known drugs, components, and excipients are used in nanoformulations. Recent data also suggest that, despite decades of research directed at hiding nanocarriers from the immune recognition, the immune system’s inherent property of clearing particulate materials can be leveraged to improve the therapeutic efficacy of drugs formulated using nanoparticles. Herein, I review current knowledge about nanoparticles’ interaction with the immune system and how these interactions contribute to nanotechnology-formulated drug products’ safety and efficacy through the lens of over a decade of nanoparticle characterization at the Nanotechnology Characterization Laboratory.
Collapse
|
156
|
Chen T, Pan F, Luo G, Jiang K, Wang H, Ding T, Li W, Zhan C, Wei X. Morphology-driven protein corona manipulation for preferential delivery of lipid nanodiscs. NANO TODAY 2022; 46:101609. [DOI: 10.1016/j.nantod.2022.101609] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
|
157
|
Ibrahim M, Ramadan E, Elsadek NE, Emam SE, Shimizu T, Ando H, Ishima Y, Elgarhy OH, Sarhan HA, Hussein AK, Ishida T. Polyethylene glycol (PEG): The nature, immunogenicity, and role in the hypersensitivity of PEGylated products. J Control Release 2022; 351:215-230. [PMID: 36165835 DOI: 10.1016/j.jconrel.2022.09.031] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/29/2022]
Abstract
Polyethylene glycol (PEG) is a versatile polymer that is widely used as an additive in foods and cosmetics, and as a carrier in PEGylated therapeutics. Even though PEG is thought to be less immunogenic, or perhaps even non-immunogenic, with a variety of physicochemical properties, there is mounting evidence that PEG causes immunogenic responses when conjugated with other materials such as proteins and nanocarriers. Under these conditions, PEG with other materials can result in the production of anti-PEG antibodies after administration. The antibodies that are induced seem to have a deleterious impact on the therapeutic efficacy of subsequently administered PEGylated formulations. In addition, hypersensitivity to PEGylated formulations could be a significant barrier to the utility of PEGylated products. Several reports have linked the presence of anti-PEG antibodies to incidences of complement activation-related pseudoallergy (CARPA) following the administration of PEGylated formulations. The use of COVID-19 mRNA vaccines, which are composed mainly of PEGylated lipid nanoparticles (LNPs), has recently gained wide acceptance, although many cases of post-vaccination hypersensitivity have been documented. Therefore, our review focuses not only on the importance of PEGs and its great role in improving the therapeutic efficacy of various medications, but also on the hypersensitivity reactions attributed to the use of PEGylated products that include PEG-based mRNA COVID-19 vaccines.
Collapse
Affiliation(s)
- Mohamed Ibrahim
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Eslam Ramadan
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Nehal E Elsadek
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Sherif E Emam
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Omar Helmy Elgarhy
- Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Hatem A Sarhan
- Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Amal K Hussein
- Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan.
| |
Collapse
|
158
|
Zalba S, Ten Hagen TLM, Burgui C, Garrido MJ. Stealth nanoparticles in oncology: Facing the PEG dilemma. J Control Release 2022; 351:22-36. [PMID: 36087801 DOI: 10.1016/j.jconrel.2022.09.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022]
Abstract
Nanoparticles (Nps) have revolutionized the landscape of many treatments, by modifying not only pharmacokinetic properties of the encapsulated agent, but also providing a significant protection of the drug from non-desired interactions, and reducing side-effects of the enclosed therapeutic, enabling co-encapsulation of possibly synergistic compounds or activities, allowing a controlled release of content and improving the therapeutic effect. Nevertheless, in systemic circulation, Nps suffer a rapid removal by opsonisation and the action of Mononuclear phagocyte system (MPS). To overcome this problem, different polymers, in particular Polyethyleneglycol (PEG), have been used to cover the surface of these nanocarriers forming a hydrophilic layer that allows the delay of the removal. These advantages contrast with some drawbacks such as the difficulty to interact with cell membranes and the development of immunological reactions, conforming the known, "PEG dilemma". To address and minimize this phenomenon, different strategies have been applied. Therefore, this review aims to summarize the state of the art of Pegylation strategies, comment in depth on the principal characteristics of PEG and describe the main alternatives, which are the use of cleavable PEG, addition of different polymers or even use other derivatives of cell membranes to camouflage Nps.
Collapse
Affiliation(s)
- Sara Zalba
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy & Nutrition, University of Navarra; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology, and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carmen Burgui
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy & Nutrition, University of Navarra
| | - María J Garrido
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy & Nutrition, University of Navarra; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
159
|
Wang H, Lin S, Wang S, Jiang Z, Ding T, Wei X, Lu Y, Yang F, Zhan C. Folic Acid Enables Targeting Delivery of Lipodiscs by Circumventing IgM-Mediated Opsonization. NANO LETTERS 2022; 22:6516-6522. [PMID: 35943299 DOI: 10.1021/acs.nanolett.2c01509] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Folic acid (FA) is one of the most widely utilized small-molecule ligands for cancer targeted drug delivery. Natural IgM was recently found to avidly absorb on the surface of FA-functionalized liposomes (FA-sLip), negatively regulating the in vivo performance by efficiently activating complement. Herein, FA-functionalized lipodiscs (FA-Disc) were constructed to successfully circumvent IgM-mediated opsonization and retained binding activity with folate receptors in vivo. The FA moiety along with the bound IgM was restricted to the highly curved rim of lipodiscs, leading to IgM incapability of presenting the membrane-bound conformation to trigger complement activation. The C1q docking, C3 binding, and C5a release were blocked and accelerated blood clearance phenomenon was mitigated of FA-Disc. FA-Disc retained folate binding activity and could effectively target folate receptor positive tumors in vivo. The present study provides a useful solution to avoid the negative regulation by IgM and achieve FA-enabled targeting by exploring disc-shaped nanocarriers.
Collapse
Affiliation(s)
- Huan Wang
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Shiqi Lin
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Songli Wang
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Zhuxuan Jiang
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Tianhao Ding
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Xiaoli Wei
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Ying Lu
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
| | - Feng Yang
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
| | - Changyou Zhan
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| |
Collapse
|
160
|
Chen H, Zhang Q. Polyglycerol fatty acid esters as alternatives to PEGylated lipids for liposome coating. Nanomedicine (Lond) 2022; 17:1027-1035. [PMID: 36000357 DOI: 10.2217/nnm-2022-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Polyglycerol (PG) is a type of biocompatible hydrophilic polyether polyol, and it is considered as a potential alternative to polyethylene glycol (PEG) in modifying nanomedicines. Materials & methods: Polyglycerol fatty acid esters (PGFEs) were modified onto liposomes and their serum stability, pharmacokinetics, in vivo distribution and the capacity to induce anti-PEG IgM were compared with PEGylated liposomes (PEG-Lips). Results: Polyglycerol 10-monostearate (PG-10-MS) displayed considerable serum stability and compatibility with mice red blood cells, and it significantly prolonged the blood circulation of liposomes in the pharmacokinetics study compared with the unmodified liposomes, with a similar biodistribution pattern to that of the PEG-Lips. Moreover, PGFE-modified liposomes were less likely to induce the production of anti-PEG IgM. Conclusion: PGFEs could be considered as good candidates to replace PEG lipids for the preparation of liposomes.
Collapse
Affiliation(s)
- Huali Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Qianyu Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
161
|
Nasra S, Bhatia D, Kumar A. Recent advances in nanoparticle-based drug delivery systems for rheumatoid arthritis treatment. NANOSCALE ADVANCES 2022; 4:3479-3494. [PMID: 36134349 PMCID: PMC9400644 DOI: 10.1039/d2na00229a] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/19/2022] [Indexed: 05/28/2023]
Abstract
Nanotechnology has increasingly emerged as a promising tool for exploring new approaches, from treating complex conditions to early detection of the onset of multiple disease states. Tailored designer nanoparticles can now more comprehensively interact with their cellular targets and various pathogens due to a similar size range and tunable surface properties. The basic goal of drug delivery is to employ pharmaceuticals only where they are needed, with as few adverse effects and off-target consequences as possible. Rheumatoid arthritis (RA) is a chronic inflammatory illness that leads to progressive loss of bone and cartilage, resulting in acute impairment, decreased life expectancy, and increased death rates. Recent advancements in treatment have significantly slowed the progression of the disease and improved the lives of many RA sufferers. Some patients, on the other hand, attain or maintain illness remission without needing to continue immunosuppressive therapy. Furthermore, a large percentage of patients do not respond to current treatments or acquire tolerance to them. As a result, novel medication options for RA therapy are still needed. Nanocarriers, unlike standard medications, are fabricated to transport drugs directly to the location of joint inflammation, evading systemic and negative effects. As a result, researchers are reconsidering medicines that were previously thought to be too hazardous for systemic delivery. This article gives an overview of contemporary nanotechnology-based tactics for treating rheumatoid arthritis, as well as how the nanotherapeutic regimen could be enhanced in the future.
Collapse
Affiliation(s)
- Simran Nasra
- Biological & Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus Navrangpura Ahmedabad Gujarat India +91796191127
| | - Dhiraj Bhatia
- Biological Engineering Discipline, Indian Institute of Technology, IIT Gandhinagar Palaj 382355 Gujarat India
| | - Ashutosh Kumar
- Biological & Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus Navrangpura Ahmedabad Gujarat India +91796191127
| |
Collapse
|
162
|
Wang S, Cheng K, Chen K, Xu C, Ma P, Dang G, Yang Y, Lei Q, Huang H, Yu Y, Fang Y, Tang Q, Jiang N, Miao H, Liu F, Zhao X, Li N. Nanoparticle-based medicines in clinical cancer therapy. NANO TODAY 2022; 45:101512. [DOI: 10.1016/j.nantod.2022.101512] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
|
163
|
Pisani S, Chiesa E, Genta I, Dorati R, Gregorini M, Grignano MA, Ramus M, Ceccarelli G, Croce S, Valsecchi C, Monti M, Rampino T, Conti B. Liposome Formulation and In Vitro Testing in Non-Physiological Conditions Addressed to Ex Vivo Kidney Perfusion. Int J Mol Sci 2022; 23:ijms23147999. [PMID: 35887348 PMCID: PMC9324182 DOI: 10.3390/ijms23147999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 02/01/2023] Open
Abstract
This work focuses on formulating liposomes to be used in isolated kidney dynamic machine perfusion in hypothermic conditions as drug delivery systems to improve preservation of transplantable organs. The need mainly arises from use of kidneys from marginal donors for transplantation that are more exposed to ischemic/reperfusion injury compared to those from standard donors. Two liposome preparation techniques, thin film hydration and microfluidic techniques, are explored for formulating liposomes loaded with two model proteins, myoglobin and bovine serum albumin. The protein-loaded liposomes are characterized for their size by DLS and morphology by TEM. Protein releases from the liposomes are tested in PERF-GEN perfusion fluid, 4 °C, and compared to the in vitro protein release in PBS, 37 °C. Fluorescent liposome uptake is analyzed by fluorescent microscope in vitro on epithelial tubular renal cell cultures and ex vivo on isolated pig kidney in hypothermic perfusion conditions. The results show that microfluidics are a superior technique for obtaining reproducible spherical liposomes with suitable size below 200 nm. Protein encapsulation efficiency is affected by its molecular weight and isoelectric point. Lowering incubation temperature slows down the proteins release; the perfusion fluid significantly affects the release of proteins sensitive to ionic media (such as BSA). Liposomes are taken up by epithelial tubular renal cells in two hours’ incubation time.
Collapse
Affiliation(s)
- Silvia Pisani
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Enrica Chiesa
- Department of Drug Sciences, University of Pavia, Vle Taramelli 12, 27100 Pavia, Italy; (E.C.); (I.G.); (R.D.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Vle Taramelli 12, 27100 Pavia, Italy; (E.C.); (I.G.); (R.D.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, Vle Taramelli 12, 27100 Pavia, Italy; (E.C.); (I.G.); (R.D.)
| | - Marilena Gregorini
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy;
| | - Maria Antonietta Grignano
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.R.); (T.R.)
| | - Marina Ramus
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.R.); (T.R.)
| | - Gabriele Ceccarelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy;
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
- Immunology & Transplantation Laboratory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.C.); (C.V.)
| | - Stefania Croce
- Immunology & Transplantation Laboratory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.C.); (C.V.)
| | - Chiara Valsecchi
- Immunology & Transplantation Laboratory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.C.); (C.V.)
| | - Manuela Monti
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, Histology and Embryology Unit University of Pavia, Biotechnology Laboratories Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Teresa Rampino
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.R.); (T.R.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Vle Taramelli 12, 27100 Pavia, Italy; (E.C.); (I.G.); (R.D.)
- Correspondence: ; Tel.: +39-0382987378
| |
Collapse
|
164
|
Tang H, Xie Y, Zhu M, Jia J, Liu R, Shen Y, Zheng Y, Guo X, Miao D, Pei J. Estrone-Conjugated PEGylated Liposome Co-Loaded Paclitaxel and Carboplatin Improve Anti-Tumor Efficacy in Ovarian Cancer and Reduce Acute Toxicity of Chemo-Drugs. Int J Nanomedicine 2022; 17:3013-3041. [PMID: 35836838 PMCID: PMC9274295 DOI: 10.2147/ijn.s362263] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/27/2022] [Indexed: 12/29/2022] Open
Abstract
Purpose Ovarian cancer is the most lethal gynecologic malignancy. The combination of paclitaxel (PTX) and carboplatin (CBP) is the first-line remedy for clinical ovarian cancer. However, due to the limitations of adverse reaction and lacking of targeting ability, the chemotherapy of ovarian cancer is still poorly effective. Here, a novel estrone (ES)-conjugated PEGylated liposome co-loaded PTX and CBP (ES-PEG-Lip-PTX/CBP) was designed for overcoming the above disadvantages. Methods ES-PEG-Lip-PTX/CBP was prepared by film hydration method and could recognize estrogen receptor (ER) over-expressing on the surface of SKOV-3 cells. The characterizations, stability and in vitro release of ES-PEG-Lip-PTX/CBP were studied. In vitro cellular uptake and its mechanism were observed by fluorescence microscope. In vivo targeting effect in tumor-bearing mice was determined. Pharmacokinetics and biodistribution were studied in ICR mice. In vitro cytotoxicity and in vivo anti-tumor efficacy were evaluated on SKOV-3 cells and tumor-bearing mice, respectively. Finally, the acute toxicity in ICR mice was explored for assessing the preliminary safety of ES-PEG-Lip-PTX/CBP. Results Our results showed that ES-PEG-Lip-PTX/CBP was spherical shape without aggregation. ES-PEG-Lip-PTX/CBP exhibited the optimum targeting effect on uptake in vitro and in vivo. The pharmacokinetics demonstrated ES-PEG-Lip-PTX/CBP had improved the pharmacokinetic behavior. In vitro cytotoxicity showed that ES-PEG-Lip-PTX/CBP maximally inhibited SKOV-3 cell proliferation and its IC50 values was 1.6 times lower than that of non-ES conjugated liposomes at 72 h. The in vivo anti-tumor efficacy study demonstrated that ES-PEG-Lip-PTX/CBP could lead strong SKOV-3 tumor growth suppression with a tumor volume inhibitory rate of 81.8%. Meanwhile, acute toxicity studies confirmed that ES-PEG-Lip-PTX/CBP significantly reduced the toxicity of the chemo drugs. Conclusion ES-PEG-Lip-PTX/CBP was successfully prepared with an optimal physicochemical and ER targeting property. The data of pharmacokinetics, anti-tumor efficacy and safety study indicated that ES-PEG-Lip-PTX/CBP could become a promising therapeutic formulation for human ovarian cancer in the future clinic.
Collapse
Affiliation(s)
- Huan Tang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Yizhuo Xie
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Ming Zhu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Juan Jia
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Rui Liu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Yujia Shen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Yucui Zheng
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Xin Guo
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Dongfanghui Miao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
165
|
Formulation and therapeutic efficacy of PEG-liposomes of sorafenib for the production of NL-PEG-SOR FUM and NL-PEG-SOR TOS. RESEARCH ON CHEMICAL INTERMEDIATES 2022. [DOI: 10.1007/s11164-022-04777-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
166
|
Luxi N, Giovanazzi A, Arcolaci A, Bonadonna P, Crivellaro MA, Cutroneo PM, Ferrajolo C, Furci F, Guidolin L, Moretti U, Olivieri E, Petrelli G, Zanoni G, Senna G, Trifirò G. Allergic Reactions to COVID-19 Vaccines: Risk Factors, Frequency, Mechanisms and Management. BioDrugs 2022; 36:443-458. [PMID: 35696066 PMCID: PMC9190452 DOI: 10.1007/s40259-022-00536-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/01/2022]
Abstract
Conventional vaccines have been widely studied, along with their risk of causing allergic reactions. These generally consist of mild local reactions and only rarely severe anaphylaxis. Although all the current COVID-19 vaccines marketed in Europe have been shown to be safe overall in the general population, early post-marketing evidence has shown that mRNA-based vaccines using novel platforms (i.e., lipid nanoparticles) were associated with an increased risk of severe allergic reactions as compared to conventional vaccines. In this paper we performed an updated literature review on frequency, risk factors, and underlying mechanisms of COVID-19 vaccine-related allergies by searching MEDLINE and Google Scholar databases. We also conducted a qualitative search on VigiBase and EudraVigilance databases to identify reports of "Hypersensitivity" and "Anaphylactic reaction" potentially related to COVID-19 vaccines (Comirnaty, Spikevax, Vaxzevria and COVID-19 Janssen Vaccine), and in EudraVigilance to estimate the reporting rates of "Anaphylactic reaction" and "Anaphylactic shock" after COVID-19 vaccination in the European population. We also summarized the scientific societies' and regulatory agencies' recommendations for prevention and management of COVID-19 vaccine-related allergic reactions, especially in those with a history of allergy.
Collapse
Affiliation(s)
- Nicoletta Luxi
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| | - Alexia Giovanazzi
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| | - Alessandra Arcolaci
- Immunology Unit, University Hospital, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Patrizia Bonadonna
- Asthma Centre and Allergy Unit, University of Verona and Verona University Hospital, Verona, Italy
| | - Maria Angiola Crivellaro
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Paola Maria Cutroneo
- Sicilian Regional Pharmacovigilance Centre, University Hospital of Messina, Messina, Italy
| | - Carmen Ferrajolo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fabiana Furci
- Immunology Unit, University Hospital, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Lucia Guidolin
- Asthma Centre and Allergy Unit, University of Verona and Verona University Hospital, Verona, Italy
| | - Ugo Moretti
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| | - Elisa Olivieri
- Asthma Centre and Allergy Unit, University of Verona and Verona University Hospital, Verona, Italy
| | - Giuliana Petrelli
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| | - Giovanna Zanoni
- Immunology Unit, University Hospital, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Gianenrico Senna
- Asthma Centre and Allergy Unit, University of Verona and Verona University Hospital, Verona, Italy
| | - Gianluca Trifirò
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Verona, Italy.
| |
Collapse
|
167
|
Mahler V, Junker AC. Anaphylaxis to additives in vaccines. ALLERGO JOURNAL INTERNATIONAL 2022; 31:123-136. [PMID: 35729887 PMCID: PMC9194891 DOI: 10.1007/s40629-022-00215-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/01/2022] [Indexed: 11/28/2022]
Abstract
SummaryAnaphylaxis in connection with the administration of vaccines occurs only very rarely. Triggers of immunoglobulin IgE-mediated and non-IgE-mediated anaphylaxis—in addition to the active ingredient itself—may be excipients contained in the vaccine due to their special properties. Some of the excipients in medicinal products are the same compounds used as additives in food. Furthermore, residues from the manufacturing process (e.g., chicken egg white, casein, antibiotics, formaldehyde) or contaminants (e.g., from the primary packaging material) may be potential triggers of anaphylaxis in vaccines. This review article provides an overview of ingredients in vaccines that pose an allergenic risk potential. The components of COVID-19 vaccines approved and marketed in Germany are discussed with regard to their potential for triggering anaphylaxis and possible pathophysiological mechanisms involved.
Collapse
Affiliation(s)
- Vera Mahler
- Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51–59, 63225 Langen, Germany
| | | |
Collapse
|
168
|
Mahler V, Junker AC. Anaphylaxie auf Zusatzstoffe in Impfstoffen. ALLERGO JOURNAL 2022; 31:22-39. [PMID: 35911653 PMCID: PMC9309021 DOI: 10.1007/s15007-022-5054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Anaphylaxien im zeitlichen Zusammenhang mit der Verabreichung von Impfstoffen treten nur sehr selten auf. Auslöser von IgE-vermittelten und nicht IgE-vermittelten Anaphylaxien können neben dem Wirkstoff selbst Hilfsstoffe sein, die aufgrund spezieller Eigenschaften im Impfstoff enthalten sind. Bei den Hilfsstoffen in Arzneimitteln handelt es sich teilweise um dieselben Verbindungen, die als Zusatzstoffe in Lebensmitteln eingesetzt werden. Weiterhin kommen in Impfstoffen Rückstände aus dem Herstellungsprozess (z. B. Hühnereiweiß, Kasein, Antibiotika, Formaldehyd) oder Kontaminationen (z. B. aus dem Primärpackmittel) als potenzielle Auslöser von Anaphylaxien in Frage. Der vorliegende Review gibt einen Überblick über Bestandteile in Impfstoffen, von denen ein allergenes Risikopotenzial ausgeht. Die Bestandteile der in Deutschland zugelassenen COVID-19-Impfstoffe als potenzielle Auslöser von Anaphylaxien und mögliche pathophysiologische Mechanismen werden diskutiert. Zitierweise: Mahler V, Junker AC. Anaphylaxis to additives in vaccines. Allergo J Int 2022;31: 123-36 https://doi.org/10.1007/s40629-022-00215-8
Collapse
Affiliation(s)
- Vera Mahler
- Leitung Abt. Allergologie, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - Ann-Christine Junker
- Leitung Abt. Allergologie, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| |
Collapse
|
169
|
Hald Albertsen C, Kulkarni JA, Witzigmann D, Lind M, Petersson K, Simonsen JB. The role of lipid components in lipid nanoparticles for vaccines and gene therapy. Adv Drug Deliv Rev 2022; 188:114416. [PMID: 35787388 PMCID: PMC9250827 DOI: 10.1016/j.addr.2022.114416] [Citation(s) in RCA: 421] [Impact Index Per Article: 140.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/03/2022] [Accepted: 06/28/2022] [Indexed: 12/21/2022]
Abstract
Lipid nanoparticles (LNPs) play an important role in mRNA vaccines against COVID-19. In addition, many preclinical and clinical studies, including the siRNA-LNP product, Onpattro®, highlight that LNPs unlock the potential of nucleic acid-based therapies and vaccines. To understand what is key to the success of LNPs, we need to understand the role of the building blocks that constitute them. In this Review, we discuss what each lipid component adds to the LNP delivery platform in terms of size, structure, stability, apparent pKa, nucleic acid encapsulation efficiency, cellular uptake, and endosomal escape. To explore this, we present findings from the liposome field as well as from landmark and recent articles in the LNP literature. We also discuss challenges and strategies related to in vitro/in vivo studies of LNPs based on fluorescence readouts, immunogenicity/reactogenicity, and LNP delivery beyond the liver. How these fundamental challenges are pursued, including what lipid components are added and combined, will likely determine the scope of LNP-based gene therapies and vaccines for treating various diseases.
Collapse
Affiliation(s)
- Camilla Hald Albertsen
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | - Jayesh A Kulkarni
- NanoVation Therapeutics Inc., 2405 Wesbrook Mall, 4th Floor, Vancouver BC V6T 1Z3, Canada
| | - Dominik Witzigmann
- NanoVation Therapeutics Inc., 2405 Wesbrook Mall, 4th Floor, Vancouver BC V6T 1Z3, Canada
| | - Marianne Lind
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | - Karsten Petersson
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | - Jens B Simonsen
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark.
| |
Collapse
|
170
|
Azlyna ASN, Ahmad S, Husna SMN, Sarmiento ME, Acosta A, Norazmi MN, Mohamud R, Kadir R. Review: Liposomes in the prophylaxis and treatment of infectious diseases. Life Sci 2022; 305:120734. [PMID: 35760094 DOI: 10.1016/j.lfs.2022.120734] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 06/08/2022] [Accepted: 06/22/2022] [Indexed: 11/15/2022]
Abstract
Infectious diseases remain as one of the major burdens among health communities as well as in the general public despite the advances in prevention and treatment. Although vaccination and vector eliminations have greatly prevented the transmission of these diseases, the effectiveness of these strategies is no longer guaranteed as new challenges such as drug resistance and toxicity as well as the missing effective therapeutics arise. Hence, the development of new tools to manage these challenges is anticipated, in which nano technology using liposomes as effective nanostructure is highly considered. In this review, we concentrate on the advantages of liposomes in the drug delivery system and the development of vaccine in the treatment of three major infectious diseases; tuberculosis (TB), malaria and HIV.
Collapse
Affiliation(s)
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Siti Muhamad Nur Husna
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Maria E Sarmiento
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Armando Acosta
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Mohd Nor Norazmi
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
171
|
Angulo C, Sanchez V, Delgado K, Monreal-Escalante E, Hernández-Adame L, Angulo M, Tello-Olea M, Reyes-Becerril M. Oral organic nanovaccines against bacterial and viral diseases. Microb Pathog 2022; 169:105648. [PMID: 35728750 DOI: 10.1016/j.micpath.2022.105648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/12/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023]
Abstract
Vaccines have saved millions of humans and animals from deadly diseases. Many vaccines are still under development to fight against lethal diseases. Indeed, subunit vaccines are a versatile approach with several advantageous attributes, but they lack strong immunogenicity. Nanotechnology is an avenue to vaccine development because nanoparticles may serve as nanocarriers and adjuvants, which are critical aspects for oral vaccines. This review provides an update of oral organic nanovaccines, describing suitable nanomaterials for oral vaccine design and recent (last five-year view) oral nanovaccine developments to fight against those principal pathogens causing human and animal diseases.
Collapse
Affiliation(s)
- Carlos Angulo
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico.
| | - Veronica Sanchez
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Karen Delgado
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Elizabeth Monreal-Escalante
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico; Cátedras-CONACYT. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Luis Hernández-Adame
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico; Cátedras-CONACYT. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Miriam Angulo
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Marlene Tello-Olea
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Martha Reyes-Becerril
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| |
Collapse
|
172
|
Padín-González E, Lancaster P, Bottini M, Gasco P, Tran L, Fadeel B, Wilkins T, Monopoli MP. Understanding the Role and Impact of Poly (Ethylene Glycol) (PEG) on Nanoparticle Formulation: Implications for COVID-19 Vaccines. Front Bioeng Biotechnol 2022; 10:882363. [PMID: 35747492 PMCID: PMC9209764 DOI: 10.3389/fbioe.2022.882363] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/09/2022] [Indexed: 12/27/2022] Open
Abstract
Poly (ethylene glycol) (PEG) is a widely used polymer in a variety of consumer products and in medicine. PEGylation refers to the conjugation of PEG to drugs or nanoparticles to increase circulation time and reduce unwanted host responses. PEG is viewed as being well-tolerated, but previous studies have identified anti-PEG antibodies and so-called pseudoallergic reactions in certain individuals. The increased use of nanoparticles as contrast agents or in drug delivery, along with the introduction of mRNA vaccines encapsulated in PEGylated lipid nanoparticles has brought this issue to the fore. Thus, while these vaccines have proven to be remarkably effective, rare cases of anaphylaxis have been reported, and this has been tentatively ascribed to the PEGylated carriers, which may trigger complement activation in susceptible individuals. Here, we provide a general overview of the use of PEGylated nanoparticles for pharmaceutical applications, and we discuss the activation of the complement cascade that might be caused by PEGylated nanomedicines for a better understanding of these immunological adverse reactions.
Collapse
Affiliation(s)
| | - Pearl Lancaster
- Department of Chemistry, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Lang Tran
- Institute of Occupational Medicine, Edinburgh, United Kingdom
| | - Bengt Fadeel
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Terence Wilkins
- School of Chemical and Process Engineering, University of Leeds, Leeds, United Kingdom
- Correspondence: Terence Wilkins, ; Marco P. Monopoli,
| | - Marco P. Monopoli
- Department of Chemistry, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
- Correspondence: Terence Wilkins, ; Marco P. Monopoli,
| |
Collapse
|
173
|
Al-Zoubi MS, Al-Zoubi RM. Nanomedicine tactics in cancer treatment: Challenge and hope. Crit Rev Oncol Hematol 2022; 174:103677. [PMID: 35385774 DOI: 10.1016/j.critrevonc.2022.103677] [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: 01/23/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 10/18/2022] Open
Abstract
Defeating cancer is the ultimate challenge and goal of oncologists, facing various obstacles along with finding effective anti-cancer therapies and understanding drug delivery mechanisms. Additionally, the translation of the experimental findings to the clinical outcomes such as specificity, delivery, toxicity, clearance, and bioavailability is another health concern. Nanomedicine is a branch of nanotechnology that has been drastically developed in the last decades. Due to the fact of various nanomaterial formulas, different nanomedicine drug delivery tactics have been developed as anti-cancer therapies. The most effective and less toxic approaches involved the active targeting drug delivery tactic, which relies on the recognition of the drug nanoparticle carriers and the cell surface marker. Accordingly, FDA approved such a group of nanomedicine drug delivery systems while other formulas are still under the clinical trial phases. Nanomedicine is showing a bright future in the treatment of cancer. Oncologists learned from cancer research the possible drug resistance that could be developed. Consequently, researchers need to be prepared for the possible adverse effect of the nanomedicine approach.
Collapse
Affiliation(s)
- Mazhar Salim Al-Zoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Raed M Al-Zoubi
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar; Department of Chemistry, Jordan University of Science and Technology, Irbid 22110, Jordan; Department of Biomedical Sciences, QU-Health, College of Health Sciences, Qatar University, Doha 2713, Qatar.
| |
Collapse
|
174
|
Mesenchymal stem cells: A living carrier for active tumor-targeted delivery. Adv Drug Deliv Rev 2022; 185:114300. [PMID: 35447165 DOI: 10.1016/j.addr.2022.114300] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/22/2022] [Accepted: 04/12/2022] [Indexed: 12/16/2022]
Abstract
The strategy of using mesenchymal stem cells (MSCs) as a living carrier for active delivery of therapeutic agents targeting tumor sites has been attempted in a wide range of studies to validate the feasibility and efficacy for tumor treatment. This approach reveals powerful tumor targeting and tumor penetration. In addition, MSCs have been confirmed to actively participate in immunomodulation of the tumor microenvironment. Thus, MSCs are not inert delivery vehicles but have a strong impact on the fate of tumor cells. In this review, these active properties of MSCs are addressed to highlight the advantages and challenges of using MSCs for tumor-targeted delivery. In addition, some of the latest examples of using MSCs to carry a variety of anti-tumor agents for tumor-targeted therapy are summarized. Recent technologies to improve the performance and safety of this delivery strategy will be introduced. The advances, applications, and challenges summarized in this review will provide a general understanding of this promising strategy for actively delivering drugs to tumor tissues.
Collapse
|
175
|
Cao Y, Zhang S, Ma M, Zhang Y. Fluorinated PEG-PEI Coated Magnetic Nanoparticles for siRNA Delivery and CXCR4 Knockdown. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1692. [PMID: 35630915 PMCID: PMC9146302 DOI: 10.3390/nano12101692] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 02/08/2023]
Abstract
CXC chemokine receptor 4 (CXCR4) is a promising therapeutic target. Previous studies have shown that intracellular delivery of siRNA to knockdown CXCR4 expression in cancer cells is an effective therapeutic strategy. To prepare efficient magnetic nucleic acid carriers, it is now necessary to improve the endocytosis efficiency of PEGylated magnetic nanoparticles. In our work, Heptafluorobutyryl-polyethylene glycol-polyethyleneimine (FPP) was first prepared and then used to coat magnetic nanoparticles (MNPs) to obtain magnetic nanocarriers FPP@MNPs. The materials were characterized by 19 F-Nuclear Magnetic Resonance (NMR), transmission electron microscope (TEM), energy dispersive spectroscopy (EDS), and dynamic light scattering (DLS). The biosecurity of FPP@MNPs was confirmed by cell viability and apoptosis experiments. Cellular uptake of FPP@MNPs and siRNA transfection enhanced by external magnetic fields were detected by fluorescence microscopy, confocal laser microscopy, and flow cytometry. The results show that the cellular uptake efficiency of FPP@MNPs was significantly improved, and transfection efficiency reached more than 90%. The knockdown of CXCR4 on the 4 T1 cell membrane was confirmed by real-time polymerase chain reaction (RT-PCR) and flow cytometry. In conclusion, the fluorinated cationic polymer-coated magnetic nanoparticles FPP@MNPs can be loaded with siRNA to reduce CXCR4 expression as well as be expected to be efficient universal siRNA carriers.
Collapse
Affiliation(s)
- Yixiang Cao
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China;
| | - Shiyin Zhang
- Nanjing Nanoeast Biotech Co., Ltd., Nanjing 211000, China;
| | - Ming Ma
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China;
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China;
| |
Collapse
|
176
|
Markowski A, Jaromin A, Migdał P, Olczak E, Zygmunt A, Zaremba-Czogalla M, Pawlik K, Gubernator J. Design and Development of a New Type of Hybrid PLGA/Lipid Nanoparticle as an Ursolic Acid Delivery System against Pancreatic Ductal Adenocarcinoma Cells. Int J Mol Sci 2022; 23:5536. [PMID: 35628352 PMCID: PMC9143619 DOI: 10.3390/ijms23105536] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/08/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
Despite many attempts, trials, and treatment procedures, pancreatic ductal adenocarcinoma (PDAC) still ranks among the most deadly and treatment-resistant types of cancer. Hence, there is still an urgent need to develop new molecules, drugs, and therapeutic methods against PDAC. Naturally derived compounds, such as pentacyclic terpenoids, have gained attention because of their high cytotoxic activity toward pancreatic cancer cells. Ursolic acid (UA), as an example, possesses a wide anticancer activity spectrum and can potentially be a good candidate for anti-PDAC therapy. However, due to its minimal water solubility, it is necessary to prepare an optimal nano-sized vehicle to overcome the low bioavailability issue. Poly(lactic-co-glycolic acid) (PLGA) polymeric nanocarriers seem to be an essential tool for ursolic acid delivery and can overcome the lack of biological activity observed after being incorporated within liposomes. PLGA modification, with the addition of PEGylated phospholipids forming the lipid shell around the polymeric core, can provide additional beneficial properties to the designed nanocarrier. We prepared UA-loaded hybrid PLGA/lipid nanoparticles using a nanoprecipitation method and subsequently performed an MTT cytotoxicity assay for AsPC-1 and BxPC-3 cells and determined the hemolytic effect on human erythrocytes with transmission electron microscopic (TEM) visualization of the nanoparticles and their cellular uptake. Hybrid UA-loaded lipid nanoparticles were also examined in terms of their stability, coating dynamics, and ursolic acid loading. We established innovative and repeatable preparation procedures for novel hybrid nanoparticles and obtained biologically active nanocarriers for ursolic acid with an IC50 below 20 µM, with an appropriate size for intravenous dosage (around 150 nm), high homogeneity of the sample (below 0.2), satisfactory encapsulation efficiency (up to 70%) and excellent stability. The new type of hybrid UA-PLGA nanoparticles represents a further step in the development of potentially effective PDAC therapies based on novel, biologically active, and promising triterpenoids.
Collapse
Affiliation(s)
- Adam Markowski
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Paweł Migdał
- Polish Academy of Science Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114 Wrocław, Poland; (P.M.); (K.P.)
- Department of Environment Hygiene and Animal Welfare, Bee Division, Wroclaw University of Environmental and Life Sciences, Chelmońskiego 38C, 51-630 Wrocław, Poland
| | - Ewa Olczak
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Adrianna Zygmunt
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Magdalena Zaremba-Czogalla
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Krzysztof Pawlik
- Polish Academy of Science Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114 Wrocław, Poland; (P.M.); (K.P.)
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| |
Collapse
|
177
|
Costa B, Boueri B, Oliveira C, Silveira I, Ribeiro AJ. Lipoplexes and polyplexes as nucleic acids delivery nanosystems: The current state and future considerations. Expert Opin Drug Deliv 2022; 19:577-594. [DOI: 10.1080/17425247.2022.2075846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Bruno Costa
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Beatriz Boueri
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Claudia Oliveira
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Isabel Silveira
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Antonio J. Ribeiro
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| |
Collapse
|
178
|
Kong YW, Dreaden EC. PEG: Will It Come Back to You? Polyethelyne Glycol Immunogenicity, COVID Vaccines, and the Case for New PEG Derivatives and Alternatives. Front Bioeng Biotechnol 2022; 10:879988. [PMID: 35573237 PMCID: PMC9092184 DOI: 10.3389/fbioe.2022.879988] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yi Wen Kong
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States
- *Correspondence: Yi Wen Kong, ; Erik C Dreaden, ,
| | - Erik C Dreaden
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- *Correspondence: Yi Wen Kong, ; Erik C Dreaden, ,
| |
Collapse
|
179
|
McNamara RP, Eason AB, Zhou Y, Bigi R, Griffith JD, Costantini LM, Rudek MA, Anders NM, Damania BA, Dittmer DP. Exosome-Encased Nucleic Acid Scaffold Chemotherapeutic Agents for Superior Anti-Tumor and Anti-Angiogenesis Activity. ACS BIO & MED CHEM AU 2022; 2:140-149. [PMID: 35480227 PMCID: PMC9026271 DOI: 10.1021/acsbiomedchemau.1c00030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022]
Abstract
Extracellular vesicles (EVs), or exosomes, play a pivotal role in tumor growth and metastasis, such as in the case of Kaposi Sarcoma. By loading tumor-derived EVs with chemotherapeutic drugs, we noted that their pro-tumor/pro-angiogenic phenotype was converted into an anti-tumor phenotype in vivo. Drug concentration in EVs was significantly higher than in clinically approved liposome formulation, as retention was facilitated by the presence of miRNAs inside the natural EVs. This demonstrates a new mechanism by which to increase the payload capacity of nanoparticles. By exploiting the targeting preferences of tumor-derived EVs, chemotherapeutics can be directed to specifically poison the cells and the microenvironment that enables metastasis.
Collapse
Affiliation(s)
- Ryan P. McNamara
- Lineberger
Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School
of Medicine, 450 West
Drive, Chapel Hill, North
Carolina 27599-9500, United States
| | - Anthony B. Eason
- Lineberger
Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School
of Medicine, 450 West
Drive, Chapel Hill, North
Carolina 27599-9500, United States
| | - Yijun Zhou
- Lineberger
Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School
of Medicine, 450 West
Drive, Chapel Hill, North
Carolina 27599-9500, United States
| | - Rachele Bigi
- Lineberger
Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School
of Medicine, 450 West
Drive, Chapel Hill, North
Carolina 27599-9500, United States
| | - Jack D. Griffith
- Lineberger
Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School
of Medicine, 450 West
Drive, Chapel Hill, North
Carolina 27599-9500, United States
| | - Lindsey M. Costantini
- Lineberger
Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School
of Medicine, 450 West
Drive, Chapel Hill, North
Carolina 27599-9500, United States
- Department
of Biological and Biomedical Sciences, North
Carolina Central University, 1801 Fayetteville Street Durham, North Carolina 27707, United States
| | - Michelle A. Rudek
- Sidney
Kimmel Comprehensive Cancer Center, Johns
Hopkins School of Medicine, 401 N. Broadway, Baltimore, Maryland 21205, United
States
| | - Nicole M. Anders
- Sidney
Kimmel Comprehensive Cancer Center, Johns
Hopkins School of Medicine, 401 N. Broadway, Baltimore, Maryland 21205, United
States
| | - Blossom A. Damania
- Lineberger
Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School
of Medicine, 450 West
Drive, Chapel Hill, North
Carolina 27599-9500, United States
| | - Dirk P. Dittmer
- Lineberger
Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School
of Medicine, 450 West
Drive, Chapel Hill, North
Carolina 27599-9500, United States
| |
Collapse
|
180
|
Application of Nanomaterials in the Prevention, Detection, and Treatment of Methicillin-Resistant Staphylococcus aureus (MRSA). Pharmaceutics 2022; 14:pharmaceutics14040805. [PMID: 35456638 PMCID: PMC9030647 DOI: 10.3390/pharmaceutics14040805] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 01/27/2023] Open
Abstract
Due to differences in geographic surveillance systems, chemical sanitization practices, and antibiotic stewardship (AS) implementation employed during the COVID-19 pandemic, many experts have expressed concerns regarding a future surge in global antimicrobial resistance (AMR). A potential beneficiary of these differences is the Gram-positive bacteria MRSA. MRSA is a bacterial pathogen with a high potential for mutational resistance, allowing it to engage various AMR mechanisms circumventing conventional antibiotic therapies and the host’s immune response. Coupled with a lack of novel FDA-approved antibiotics reaching the clinic, the onus is on researchers to develop alternative treatment tools to mitigate against an increase in pathogenic resistance. Mitigation strategies can take the form of synthetic or biomimetic nanomaterials/vesicles employed in vaccines, rapid diagnostics, antibiotic delivery, and nanotherapeutics. This review seeks to discuss the current potential of the aforementioned nanomaterials in detecting and treating MRSA.
Collapse
|
181
|
Cao Y, Dong X, Chen X. Polymer-Modified Liposomes for Drug Delivery: From Fundamentals to Applications. Pharmaceutics 2022; 14:pharmaceutics14040778. [PMID: 35456613 PMCID: PMC9026371 DOI: 10.3390/pharmaceutics14040778] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Liposomes are highly advantageous platforms for drug delivery. To improve the colloidal stability and avoid rapid uptake by the mononuclear phagocytic system of conventional liposomes while controlling the release of encapsulated agents, modification of liposomes with well-designed polymers to modulate the physiological, particularly the interfacial properties of the drug carriers, has been intensively investigated. Briefly, polymers are incorporated into liposomes mainly using “grafting” or “coating”, defined according to the configuration of polymers at the surface. Polymer-modified liposomes preserve the advantages of liposomes as drug-delivery carriers and possess specific functionality from the polymers, such as long circulation, precise targeting, and stimulus-responsiveness, thereby resulting in improved pharmacokinetics, biodistribution, toxicity, and therapeutic efficacy. In this review, we summarize the progress in polymer-modified liposomes for drug delivery, focusing on the change in physiological properties of liposomes and factors influencing the overall therapeutic efficacy.
Collapse
Affiliation(s)
- Yifeng Cao
- Department of Electronic Chemicals, Institute of Zhejiang University-Quzhou, Quzhou 324000, China
- Correspondence: (Y.C.); (X.C.)
| | - Xinyan Dong
- School of Biological and Chemical Engineering, NingboTech University, Ningbo 315100, China;
| | - Xuepeng Chen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
- Correspondence: (Y.C.); (X.C.)
| |
Collapse
|
182
|
Münter R, Stavnsbjerg C, Christensen E, Thomsen ME, Stensballe A, Hansen AE, Parhamifar L, Kristensen K, Simonsen JB, Larsen JB, Andresen TL. Unravelling Heterogeneities in Complement and Antibody Opsonization of Individual Liposomes as a Function of Surface Architecture. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106529. [PMID: 35187804 DOI: 10.1002/smll.202106529] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/24/2022] [Indexed: 06/14/2023]
Abstract
Coating nanoparticles with poly(ethylene glycol) (PEG) is widely used to achieve long-circulating properties after infusion. While PEG reduces binding of opsonins to the particle surface, immunogenic anti-PEG side-effects show that PEGylated nanoparticles are not truly "stealth" to surface active proteins. A major obstacle for understanding the complex interplay between opsonins and nanoparticles is the averaging effects of the bulk assays that are typically applied to study protein adsorption to nanoparticles. Here, a microscopy-based method for directly quantifying opsonization at the single nanoparticle level is presented. Various surface coatings are investigated on liposomes, including PEG, and show that opsonization by both antibodies and complement C3b is highly dependent on the surface chemistry. It is further demonstrated that this opsonization is heterogeneous, with opsonized and non-opsonized liposomes co-existing in the same ensemble. Surface coatings modify the percentage of opsonized liposomes and/or opsonin surface density on the liposomes, with strikingly different patterns for antibodies and complement. Thus, this assay provides mechanistic details about opsonization at the single nanoparticle level previously inaccessible to established bulk assays.
Collapse
Affiliation(s)
- Rasmus Münter
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Camilla Stavnsbjerg
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Esben Christensen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Mikkel E Thomsen
- Department of Health Science and Technology, Aalborg University, Aalborg Ø, 9220, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg Ø, 9220, Denmark
| | - Anders E Hansen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Ladan Parhamifar
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Kasper Kristensen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Jens B Simonsen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Jannik B Larsen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Thomas L Andresen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| |
Collapse
|
183
|
Nanoformulation of plant-based natural products for type 2 diabetes mellitus: From formulation design to therapeutic applications. Curr Ther Res Clin Exp 2022; 96:100672. [PMID: 35586563 PMCID: PMC9108891 DOI: 10.1016/j.curtheres.2022.100672] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
Inorganic element based nanoformulations were prominent in the delivery drug leads. Polymer and lipid based nanoformulations are emerging as novel formulations. Majority of investigations on nanoherbal formulations were on in vitro models. Proper glycemic control was an important property in nanoherbalformulations.
Background Herbal remedies are used to manage type 2 diabetes mellitus (type 2 DM) as the sole treatment or as a complementary therapy. Limitations of herbal remedies, such as poor stability and limited absorption, impede their development as therapeutic agents, which could be overcome by nanoformulations. Objectives This review attempts to summarize the studies reported between 2009 and 2020 in the development of medicinal plant-based nanoformulations for the management of type 2 DM, discuss formulation methods, mechanisms of action, and identify gaps in the literature to conduct future research on nanoparticle-based herbal treatment options targeting type 2 DM. Methods To retrieve articles published between January 2009 and December 2020, the electronic databases PubMed, Science Direct, and Google Scholar were searched with the keywords nanoparticle, plant, and diabetes in the entire text. Peer-reviewed research articles on herbal nanoformulations published in English-language based on in vitro and/or in vivo models of type 2 DM and/or its complications were included. The literature search and selection of titles/abstracts were carried out independently by 2 authors. The list of full-text articles was selected considering inclusion and exclusion criteria, with the agreement of all the authors. Results Among the reported studies, 68% of the studies were on inorganic herbal nanoformulations, whereas 17% and 8% were of polymer-based and lipid-based herbal nanoformulations, respectively. Some of the important biological properties of nanoformulations included improvement in glycemic control and insulin levels, inhibition of the formation of advanced glycation end products, and regeneration of pancreatic β cells. The aforementioned properties were observed by screening nanoformulations using in vitro cellular and noncellular models, as well as in vivo animal models of type 2 DM studied for acute or subacute durations. Only 2 clinical trials with patients with diabetes were reported, indicating the need for further research on medicinal plant-based nanoformulations as a therapeutic option for the management of type 2 DM. Conclusions Medicinal plant extracts and isolated compounds have been nanoformulated using various methods. The properties of the nanoformulations were found superior to those of the corresponding herbal extracts and isolated compounds. At both the preclinical and clinical levels, there are a number of poorly explored research areas in the development and bioactivity assessment of herbal nanoformulations. (Curr Ther Res Clin Exp. 2022; 83:XXX–XXX) © 2022 Elsevier HS Journals, Inc.
Collapse
|
184
|
Ibarra-Sánchez LÁ, Gámez-Méndez A, Martínez-Ruiz M, Nájera-Martínez EF, Morales-Flores BA, Melchor-Martínez EM, Sosa-Hernández JE, Parra-Saldívar R, Iqbal HMN. Nanostructures for drug delivery in respiratory diseases therapeutics: Revision of current trends and its comparative analysis. J Drug Deliv Sci Technol 2022; 70:103219. [PMID: 35280919 PMCID: PMC8896872 DOI: 10.1016/j.jddst.2022.103219] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 02/02/2022] [Accepted: 02/26/2022] [Indexed: 02/08/2023]
Abstract
Respiratory diseases are leading causes of death and disability in developing and developed countries. The burden of acute and chronic respiratory diseases has been rising throughout the world and represents a major problem in the public health system. Acute respiratory diseases include pneumonia, influenza, SARS-CoV-2 and MERS viral infections; while chronic obstructive pulmonary disease (COPD), asthma and, occupational lung diseases (asbestosis, pneumoconiosis) and other parenchymal lung diseases namely lung cancer and tuberculosis are examples of chronic respiratory diseases. Importantly, chronic respiratory diseases are not curable and treatments for acute pathologies are particularly challenging. For that reason, the integration of nanotechnology to existing drugs or for the development of new treatments potentially benefits the therapeutic goals by making drugs more effective and exhibit fewer undesirable side effects to treat these conditions. Moreover, the integration of different nanostructures enables improvement of drug bioavailability, transport and delivery compared to stand-alone drugs in traditional respiratory therapy. Notably, there has been great progress in translating nanotechnology-based cancer therapies and diagnostics into the clinic; however, researchers in recent years have focused on the application of nanostructures in other relevant pulmonary diseases as revealed in our database search. Furthermore, polymeric nanoparticles and micelles are the most studied nanostructures in a wide range of diseases; however, liposomal nanostructures are recognized to be some of the most successful commercial drug delivery systems. In conclusion, this review presents an overview of the recent and relevant research in drug delivery systems for the treatment of different pulmonary diseases and outlines the trends, limitations, importance and application of nanomedicine technology in treatment and diagnosis and future work in this field.
Collapse
Affiliation(s)
- Luis Ángel Ibarra-Sánchez
- Tecnológico de Monterrey, School of Engineering and Sciences, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, N.L., Mexico
| | - Ana Gámez-Méndez
- Universidad de Monterrey, Department of Basic Sciences, Av. Ignacio Morones Prieto 4500 Pte., 66238, San Pedro Garza García, Nuevo León, Mexico
| | - Manuel Martínez-Ruiz
- Tecnológico de Monterrey, School of Engineering and Sciences, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, N.L., Mexico
| | - Erik Francisco Nájera-Martínez
- Tecnológico de Monterrey, School of Engineering and Sciences, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, N.L., Mexico
| | - Brando Alan Morales-Flores
- Tecnológico de Monterrey, School of Engineering and Sciences, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, N.L., Mexico
| | - Elda M Melchor-Martínez
- Tecnológico de Monterrey, School of Engineering and Sciences, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, N.L., Mexico
| | - Juan Eduardo Sosa-Hernández
- Tecnológico de Monterrey, School of Engineering and Sciences, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, N.L., Mexico
| | - Roberto Parra-Saldívar
- Tecnológico de Monterrey, School of Engineering and Sciences, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, N.L., Mexico
| | - Hafiz M N Iqbal
- Tecnológico de Monterrey, School of Engineering and Sciences, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, N.L., Mexico
| |
Collapse
|
185
|
Formulation and Characterization of Stimuli-Responsive Lecithin-Based Liposome Complexes with Poly(acrylic acid)/Poly(N,N-dimethylaminoethyl methacrylate) and Pluronic® Copolymers for Controlled Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14040735. [PMID: 35456569 PMCID: PMC9029292 DOI: 10.3390/pharmaceutics14040735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
Polymer–liposome complexes (PLCs) can be efficiently applied for the treatment and/or diagnosis of several types of diseases, such as cancerous, dermatological, neurological, ophthalmic and orthopedic. In this work, temperature-/pH-sensitive PLC-based systems for controlled release were developed and characterized. The selected hydrophilic polymeric setup consists of copolymers of Pluronic®-poly(acrylic acid) (PLU-PAA) and Pluronic®-poly(N,N-dimethylaminoethyl methacrylate) (PLU-PD) synthesized by atom transfer radical polymerization (ATRP). The copolymers were incorporated into liposomes formulated from soybean lecithin, with different copolymer/phospholipid ratios (2.5, 5 and 10%). PLCs were characterized by evaluating their particle size, polydispersity, surface charge, capacity of release and encapsulation efficiency. Their cytotoxic potential was assessed by determining the viability of human epithelial cells exposed to them. The results showed that the incorporation of the synthesized copolymers positively contributed to the stabilization of the liposomes. The main accomplishments of this work were the innovative synthesis of PLU-PD and PLU-PAA by ATRP, and the liposome stabilization by their incorporation. The formulated PLCs exhibited relevant characteristics, notably stimuli-responsive attributes upon slight changes in pH and/or temperature, with proven absence of cellular toxicity, which could be of interest for the treatment or diagnosis of all diseases that cause some particular pH/temperature change in the target area.
Collapse
|
186
|
Arber Raviv S, Alyan M, Egorov E, Zano A, Harush MY, Pieters C, Korach-Rechtman H, Saadya A, Kaneti G, Nudelman I, Farkash S, Flikshtain OD, Mekies LN, Koren L, Gal Y, Dor E, Shainsky J, Shklover J, Adir Y, Schroeder A. Lung targeted liposomes for treating ARDS. J Control Release 2022; 346:421-433. [PMID: 35358610 PMCID: PMC8958843 DOI: 10.1016/j.jconrel.2022.03.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 12/18/2022]
Abstract
Acute Respiratory Distress Syndrome (ARDS), associated with Covid-19 infections, is characterized by diffuse lung damage, inflammation and alveolar collapse that impairs gas exchange, leading to hypoxemia and patient’ mortality rates above 40%. Here, we describe the development and assessment of 100-nm liposomes that are tailored for pulmonary delivery for treating ARDS, as a model for lung diseases. The liposomal lipid composition (primarily DPPC) was optimized to mimic the lung surfactant composition, and the drug loading process of both methylprednisolone (MPS), a steroid, and N-acetyl cysteine (NAC), a mucolytic agent, reached an encapsulation efficiency of 98% and 92%, respectively. In vitro, treating lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages with the liposomes decreased TNFα and nitric oxide (NO) secretion, while NAC increased the penetration of nanoparticles through the mucus. In vivo, we used LPS-induced lung inflammation model to assess the accumulation and therapeutic efficacy of the liposomes in C57BL/6 mice, either by intravenous (IV), endotracheal (ET) or IV plus ET nanoparticles administrations. Using both administration methods, liposomes exhibited an increased accumulation profile in the inflamed lungs over 48 h. Interestingly, while IV-administrated liposomes distributed widely throughout the lung, ET liposomes were present in lungs parenchyma but were not detected at some distal regions of the lungs, possibly due to imperfect airflow regimes. Twenty hours after the different treatments, lungs were assessed for markers of inflammation. We found that the nanoparticle treatment had a superior therapeutic effect compared to free drugs in treating ARDS, reducing inflammation and TNFα, IL-6 and IL-1β cytokine secretion in bronchoalveolar lavage (BAL), and that the combined treatment, delivering nanoparticles IV and ET simultaneously, had the best outcome of all treatments. Interestingly, also the DPPC lipid component alone played a therapeutic role in reducing inflammatory markers in the lungs. Collectively, we show that therapeutic nanoparticles accumulate in inflamed lungs holding potential for treating lung disorders. Significance In this study we compare intravenous versus intratracheal delivery of nanoparticles for treating lung disorders, specifically, acute respiratory distress syndrome (ARDS). By co-loading two medications into lipid nanoparticles, we were able to reduce both inflammation and mucus secretion in the inflamed lungs. Both modes of delivery resulted in high nanoparticle accumulation in the lungs, intravenously administered nanoparticles reached lung endothelial while endotracheal delivery reached lung epithelial. Combining both delivery approaches simultaneously provided the best ARDS treatment outcome.
Collapse
Affiliation(s)
- Sivan Arber Raviv
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Mohammed Alyan
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel; The Interdisciplinary Program for Biotechnology, Technion, Haifa, 3200003, Israel
| | - Egor Egorov
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Agam Zano
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Moshit Yaskin Harush
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Calvin Pieters
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Hila Korach-Rechtman
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Adi Saadya
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Galoz Kaneti
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Igor Nudelman
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Shai Farkash
- Department of Pathology, Emek Medical Center, Afula, Israel
| | - Ofri Doppelt Flikshtain
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Lucy N Mekies
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Lilach Koren
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Yoav Gal
- Office Of Assistant Minister of Defense for CBRN Defense, Ministry of Defense, Tel-Aviv, Israel
| | - Ella Dor
- Office Of Assistant Minister of Defense for CBRN Defense, Ministry of Defense, Tel-Aviv, Israel
| | - Janna Shainsky
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Jeny Shklover
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Yochai Adir
- Pulmonary Division, Lady Davis, Carmel Medical Center, Faculty of Medicine, The Technion Institute of Technology, Haifa, Israel
| | - Avi Schroeder
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
187
|
Gonciar D, Mocan T, Agoston-Coldea L. Nanoparticles Targeting the Molecular Pathways of Heart Remodeling and Regeneration. Pharmaceutics 2022; 14:pharmaceutics14040711. [PMID: 35456545 PMCID: PMC9028351 DOI: 10.3390/pharmaceutics14040711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
Cardiovascular diseases are the main cause of death worldwide, a trend that will continue to grow over the next decade. The heart consists of a complex cellular network based mainly on cardiomyocytes, but also on endothelial cells, smooth muscle cells, fibroblasts, and pericytes, which closely communicate through paracrine factors and direct contact. These interactions serve as valuable targets in understanding the phenomenon of heart remodeling and regeneration. The advances in nanomedicine in the controlled delivery of active pharmacological agents are remarkable and may provide substantial contribution to the treatment of heart diseases. This review aims to summarize the main mechanisms involved in cardiac remodeling and regeneration and how they have been applied in nanomedicine.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| | - Teodora Mocan
- Physiology Department, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca 400162, Romania
- Correspondence:
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| |
Collapse
|
188
|
Liposome-Tethered Gold Nanoparticles Triggered by Pulsed NIR Light for Rapid Liposome Contents Release and Endosome Escape. Pharmaceutics 2022; 14:pharmaceutics14040701. [PMID: 35456535 PMCID: PMC9025641 DOI: 10.3390/pharmaceutics14040701] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Remote triggering of contents release with micron spatial and sub-second temporal resolution has been a long-time goal of medical and technical applications of liposomes. Liposomes can sequester a variety of bioactive water-soluble ions, ligands and enzymes, and oligonucleotides. The bilayer that separates the liposome interior from the exterior solution provides a physical barrier to contents release and degradation. Tethering plasmon-resonant, hollow gold nanoshells to the liposomes, or growing gold nanoparticles directly on the liposome exterior, allows liposome contents to be released by nanosecond or shorter pulses of near-infrared light (NIR). Gold nanoshells or nanoparticles strongly adsorb NIR light; cells, tissues, and physiological media are transparent to NIR, allowing penetration depths of millimeters to centimeters. Nano to picosecond pulses of NIR light rapidly heat the gold nanoshells, inducing the formation of vapor nanobubbles, similar to cavitation bubbles. The collapse of the nanobubbles generates mechanical forces that rupture bilayer membranes to rapidly release liposome contents at the preferred location and time. Here, we review the syntheses, characterization, and applications of liposomes coupled to plasmon-resonant gold nanostructures for delivering a variety of biologically important contents in vitro and in vivo with sub-micron spatial control and sub-second temporal control.
Collapse
|
189
|
Qiao K, Xu L, Tang J, Wang Q, Lim KS, Hooper G, Woodfield TBF, Liu G, Tian K, Zhang W, Cui X. The advances in nanomedicine for bone and cartilage repair. J Nanobiotechnology 2022; 20:141. [PMID: 35303876 PMCID: PMC8932118 DOI: 10.1186/s12951-022-01342-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/01/2022] [Indexed: 12/24/2022] Open
Abstract
With the gradual demographic shift toward an aging and obese society, an increasing number of patients are suffering from bone and cartilage injuries. However, conventional therapies are hindered by the defects of materials, failing to adequately stimulate the necessary cellular response to promote sufficient cartilage regeneration, bone remodeling and osseointegration. In recent years, the rapid development of nanomedicine has initiated a revolution in orthopedics, especially in tissue engineering and regenerative medicine, due to their capacity to effectively stimulate cellular responses on a nanoscale with enhanced drug loading efficiency, targeted capability, increased mechanical properties and improved uptake rate, resulting in an improved therapeutic effect. Therefore, a comprehensive review of advancements in nanomedicine for bone and cartilage diseases is timely and beneficial. This review firstly summarized the wide range of existing nanotechnology applications in the medical field. The progressive development of nano delivery systems in nanomedicine, including nanoparticles and biomimetic techniques, which are lacking in the current literature, is further described. More importantly, we also highlighted the research advancements of nanomedicine in bone and cartilage repair using the latest preclinical and clinical examples, and further discussed the research directions of nano-therapies in future clinical practice.
Collapse
Affiliation(s)
- Kai Qiao
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Lu Xu
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China
- Department of Dermatology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Junnan Tang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 61004, Sichuan, China
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Gary Hooper
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Guozhen Liu
- School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, 518172, Guangdong, China
| | - Kang Tian
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China.
| | - Weiguo Zhang
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China.
| | - Xiaolin Cui
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China.
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand.
| |
Collapse
|
190
|
Zoulikha M, He W. Targeted Drug Delivery for Chronic Lymphocytic Leukemia. Pharm Res 2022; 39:441-461. [DOI: 10.1007/s11095-022-03214-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/21/2022] [Indexed: 02/06/2023]
|
191
|
van der Koog L, Gandek TB, Nagelkerke A. Liposomes and Extracellular Vesicles as Drug Delivery Systems: A Comparison of Composition, Pharmacokinetics, and Functionalization. Adv Healthc Mater 2022; 11:e2100639. [PMID: 34165909 PMCID: PMC11468589 DOI: 10.1002/adhm.202100639] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/27/2021] [Indexed: 12/11/2022]
Abstract
Over the past decades, lipid-based nanoparticle drug delivery systems (DDS) have caught the attention of researchers worldwide, encouraging the field to rapidly develop improved ways for effective drug delivery. One of the most prominent examples is liposomes, which are spherical shaped artificial vesicles composed of lipid bilayers and able to encapsulate both hydrophilic and hydrophobic materials. At the same time, biological nanoparticles naturally secreted by cells, called extracellular vesicles (EVs), have emerged as promising more complex biocompatible DDS. In this review paper, the differences and similarities in the composition of both vesicles are evaluated, and critical mediators that affect their pharmacokinetics are elucidate. Different strategies that have been assessed to tweak the pharmacokinetics of both liposomes and EVs are explored, detailing the effects on circulation time, targeting capacity, and cytoplasmic delivery of therapeutic cargo. Finally, whether a hybrid system, consisting of a combination of only the critical constituents of both vesicles, could offer the best of both worlds is discussed. Through these topics, novel leads for further research are provided and, more importantly, gain insight in what the liposome field and the EV field can learn from each other.
Collapse
Affiliation(s)
- Luke van der Koog
- Molecular PharmacologyGroningen Research Institute of PharmacyGRIAC Research Institute, University Medical Center GroningenUniversity of GroningenP.O. Box 196, XB10Groningen9700 ADThe Netherlands
| | - Timea B. Gandek
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| | - Anika Nagelkerke
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| |
Collapse
|
192
|
Talkington AM, McSweeney MD, Wessler T, Rath MK, Li Z, Zhang T, Yuan H, Frank JE, Forest MG, Cao Y, Lai SK. A PBPK model recapitulates early kinetics of anti-PEG antibody-mediated clearance of PEG-liposomes. J Control Release 2022; 343:518-527. [PMID: 35066099 PMCID: PMC9080587 DOI: 10.1016/j.jconrel.2022.01.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/23/2022]
Abstract
PEGylation is routinely used to extend the systemic circulation of various protein therapeutics and nanomedicines. Nonetheless, mounting evidence is emerging that individuals exposed to select PEGylated therapeutics can develop antibodies specific to PEG, i.e., anti-PEG antibodies (APA). In turn, APA increase both the risk of hypersensitivity to the drug as well as potential loss of efficacy due to accelerated blood clearance of the drug. Despite the broad implications of APA, the timescales and systemic specificity by which APA can alter the pharmacokinetics and biodistribution of PEGylated drugs remain not well understood. Here, we developed a physiologically based pharmacokinetic (PBPK) model designed to resolve APA's impact on both early- and late-phase pharmacokinetics and biodistribution of intravenously administered PEGylated drugs. Our model accurately recapitulates PK and biodistribution data obtained from PET/CT imaging of radiolabeled PEG-liposomes and PEG-uricase in mice with and without APA, as well as serum levels of PEG-uricase in humans. Our work provides another illustration of the power of high-resolution PBPK models for understanding the pharmacokinetic impacts of anti-drug antibodies and the dynamics with which antibodies can mediate clearance of foreign species.
Collapse
Affiliation(s)
- Anne M Talkington
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Morgan D McSweeney
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy Wessler
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA; Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Marielle K Rath
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Zibo Li
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Tao Zhang
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Hong Yuan
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, UNC Chapel Hill, USA
| | | | - M Gregory Forest
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA; Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA; Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina, Chapel Hill, NC, USA
| | - Samuel K Lai
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
193
|
Ozer I, Pitoc GA, Layzer JM, Moreno A, Olson LB, Layzer KD, Hucknall AM, Sullenger BA, Chilkoti A. PEG-Like Brush Polymer Conjugate of RNA Aptamer That Shows Reversible Anticoagulant Activity and Minimal Immune Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107852. [PMID: 34994037 DOI: 10.1002/adma.202107852] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/28/2021] [Indexed: 06/14/2023]
Abstract
Ribonucleic acid (RNA) therapeutics are an emerging class of drugs. RNA aptamers are of significant therapeutic and clinical interest because their activity can be easily reversed in vivo-a useful feature that is difficult to achieve using other therapeutic modalities. Despite their therapeutic promise, RNA aptamers are limited by their poor blood circulation. The attachment of polyethylene glycol (PEG) to RNA aptamers addresses this limitation. However, an RNA aptamer-PEG conjugate that is a reversible anticoagulant fails in a clinical trial due to the reactivity of the conjugate with pre-existing PEG antibodies and has cast a pall over PEGylation of aptamers and other biologics, despite its long history of utility in drug delivery. Here, PEG antibody-reactivity of this RNA aptamer is eliminated by conjugating it to a next-generation PEG-like brush polymer-poly[(oligoethylene glycol) methyl ether methacrylate)] (POEGMA). The conjugate retained the drug's therapeutic action and the ability to be easily reversed. Importantly, this conjugate does not bind pre-existing PEG antibodies that are prevalent in humans and does not induce a humoral immune response against the polymer itself in mice. These findings suggest a path to rescuing the PEGylation of RNA therapeutics and vaccines from the deleterious side-effects of PEG.
Collapse
Affiliation(s)
- Imran Ozer
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - George A Pitoc
- Department of Surgery, Duke University Medical Center, Durham, NC, 27707, USA
| | - Juliana M Layzer
- Department of Surgery, Duke University Medical Center, Durham, NC, 27707, USA
- Duke Clinical and Translational Science Institute, Durham, NC, 27707, USA
| | - Angelo Moreno
- Department of Surgery, Duke University Medical Center, Durham, NC, 27707, USA
| | - Lyra B Olson
- Department of Surgery, Duke University Medical Center, Durham, NC, 27707, USA
| | - Kyle D Layzer
- Department of Surgery, Duke University Medical Center, Durham, NC, 27707, USA
| | - Angus M Hucknall
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Bruce A Sullenger
- Department of Surgery, Duke University Medical Center, Durham, NC, 27707, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
194
|
Lombardo D, Kiselev MA. Methods of Liposomes Preparation: Formation and Control Factors of Versatile Nanocarriers for Biomedical and Nanomedicine Application. Pharmaceutics 2022; 14:pharmaceutics14030543. [PMID: 35335920 PMCID: PMC8955843 DOI: 10.3390/pharmaceutics14030543] [Citation(s) in RCA: 197] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022] Open
Abstract
Liposomes are nano-sized spherical vesicles composed of an aqueous core surrounded by one (or more) phospholipid bilayer shells. Owing to their high biocompatibility, chemical composition variability, and ease of preparation, as well as their large variety of structural properties, liposomes have been employed in a large variety of nanomedicine and biomedical applications, including nanocarriers for drug delivery, in nutraceutical fields, for immunoassays, clinical diagnostics, tissue engineering, and theranostics formulations. Particularly important is the role of liposomes in drug-delivery applications, as they improve the performance of the encapsulated drugs, reducing side effects and toxicity by enhancing its in vitro- and in vivo-controlled delivery and activity. These applications stimulated a great effort for the scale-up of the formation processes in view of suitable industrial development. Despite the improvements of conventional approaches and the development of novel routes of liposome preparation, their intrinsic sensitivity to mechanical and chemical actions is responsible for some critical issues connected with a limited colloidal stability and reduced entrapment efficiency of cargo molecules. This article analyzes the main features of the formation and fabrication techniques of liposome nanocarriers, with a special focus on the structure, parameters, and the critical factors that influence the development of a suitable and stable formulation. Recent developments and new methods for liposome preparation are also discussed, with the objective of updating the reader and providing future directions for research and development.
Collapse
Affiliation(s)
- Domenico Lombardo
- Consiglio Nazionale delle Ricerche, Istituto per i Processi Chimico-Fisici, 98158 Messina, Italy
- Correspondence: ; Tel.: +39-090-39762222
| | - Mikhail A. Kiselev
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, 141980 Dubna, Moscow Region, Russia;
- Department of Nuclear Physics, Dubna State University, 141980 Dubna, Moscow Region, Russia
- Physics Department, Lomonosov Moscow State University, 119991 Moscow, Moscow Region, Russia
| |
Collapse
|
195
|
Jacob S, Nair AB, Shah J, Gupta S, Boddu SHS, Sreeharsha N, Joseph A, Shinu P, Morsy MA. Lipid Nanoparticles as a Promising Drug Delivery Carrier for Topical Ocular Therapy-An Overview on Recent Advances. Pharmaceutics 2022; 14:533. [PMID: 35335909 PMCID: PMC8955373 DOI: 10.3390/pharmaceutics14030533] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/16/2022] Open
Abstract
Due to complicated anatomical and physical properties, targeted drug delivery to ocular tissues continues to be a key challenge for formulation scientists. Various attempts are currently being made to improve the in vivo performance of therapeutic molecules by encapsulating them in various nanocarrier systems or devices and administering them via invasive/non-invasive or minimally invasive drug administration methods. Biocompatible and biodegradable lipid nanoparticles have emerged as a potential alternative to conventional ocular drug delivery systems to overcome various ocular barriers. Lipid-based nanocarrier systems led to major technological advancements and therapeutic advantages during the last few decades of ocular therapy, such as high precorneal residence time, sustained drug release profile, minimum dosing frequency, decreased drug toxicity, targeted site delivery, and, therefore, an improvement in ocular bioavailability. In addition, such formulations can be given as fine dispersion in patient-friendly droppable preparation without causing blurred vision and ocular sensitivity reactions. The unique advantages of lipid nanoparticles, namely, solid lipid nanoparticles, nanostructured lipid carriers, nanoemulsions, and liposomes in intraocular targeted administration of various therapeutic drugs are extensively discussed. Ongoing and completed clinical trials of various liposome-based formulations and various characterization techniques designed for nanoemulsion in ocular delivery are tabulated. This review also describes diverse solid lipid nanoparticle preparation methods, procedures, advantages, and limitations. Functionalization approaches to overcome the drawbacks of lipid nanoparticles, as well as the exploration of new functional additives with the potential to improve the penetration of macromolecular pharmaceuticals, would quickly progress the challenging field of ocular drug delivery systems.
Collapse
Affiliation(s)
- Shery Jacob
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Anroop B. Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.B.N.); (N.S.); (M.A.M.)
| | - Jigar Shah
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India;
| | - Sumeet Gupta
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana 133203, India;
| | - Sai H. S. Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates;
| | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.B.N.); (N.S.); (M.A.M.)
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bangalore 560035, India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Pottathil Shinu
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | - Mohamed A. Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.B.N.); (N.S.); (M.A.M.)
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
| |
Collapse
|
196
|
Branched PEG-modification: A new strategy for nanocarriers to evade of the accelerated blood clearance phenomenon and enhance anti-tumor efficacy. Biomaterials 2022; 283:121415. [PMID: 35217484 DOI: 10.1016/j.biomaterials.2022.121415] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 01/18/2023]
Abstract
PEGylation is one of the most successful technologies for reducing immunogenicity, improving the stability and circulation time of nanocarriers, and has been applied in the clinic for over three decades. However, linear PEG-modified nanocarriers have been found to induce anti-PEG IgM at the first injection, which triggers the accelerated blood clearance (ABC) phenomenon upon repeated injections. Furthermore, clinical and research evidence has revealed that anti-PEG antibodies also cause serious complement activation-related pseudoallergies (CARPA), which greatly reduce the safety of linear PEGylated nanocarriers. In this study, as an alternative to linear PEG, branched PEG was selected owing to its low antigenicity. We pioneer the use of branched PEG lipid derivatives [DSPE-mPEG2,n (n = 2, 10, and 20 kDa)] to modify nanoemulsions (PE2,n) and liposomes (PL2,n). Upon characterization, PE2,n and PL2,n showed similar physicochemical properties to linear DSPE-mPEG2000-modified nanocarriers in terms of size, polydispersity index (PDI), and zeta potential. However, our pharmacokinetics study surprisingly indicated that PE2,n and PL2,n did not induce the ABC phenomenon after repeated injection. This may be attributed to the fact that PE2,n and PL2,n induced noticeably lower levels of anti-PEG IgM than linear PEG-modified nanocarriers and did not activate the complement system. Furthermore, we are the first to investigate the anti-tumor efficacy of DSPE-mPEG2,n-modified liposomal doxorubicin (DOX). The pharmacodynamic experiments showed that DSPE-mPEG2,n-m-modified liposomal DOX had better in vivo anti-tumor effects than linear DSPE-mPEG2000-modified liposomes. Therefore, we speculate that DSPE-mPEG2,n-modified nanocarriers possess promising prospects in avoiding the ABC phenomenon, reducing CARPA, and improving the anti-tumor efficacy of encapsulated drugs.
Collapse
|
197
|
Leng J, Dai X, Cheng X, Zhou H, Wang D, Zhao J, Ma K, Cui C, Wang L, Guo Z. Biomimetic Cucurbitacin B-Polydopamine Nanoparticles for Synergistic Chemo-Photothermal Therapy of Breast Cancer. Front Bioeng Biotechnol 2022; 10:841186. [PMID: 35223801 PMCID: PMC8864241 DOI: 10.3389/fbioe.2022.841186] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/13/2022] [Indexed: 11/23/2022] Open
Abstract
Breast cancer is the most common malignant tumor in women. Researchers have found that the combined use of multiple methods to treat tumors is a promising strategy. Here, we have developed a biomimetic nano-platform PDA@MB for tumor targeted photothermal therapy (PTT) combined with chemotherapy. The 4T1 cell membrane loaded with cucurbitacin B (CuB) was used to coat polydopamine (PDA) nanoparticles, which gave PDA@MB nanoparticles the ability to target tumors and escape immune cells from phagocytosis. PDA@MB showed excellent photothermal performance including high photothermal conversion efficiency and photostability, and exhibited outstanding in vitro PTT effect under NIR laser irradiation. The high temperature ruptured the PDA@MB membrane to release CuB, which changed the tumor hypoxic environment, down-regulated the FAK/MMP signaling pathway, and significantly inhibited the metastasis and proliferation of tumor cells. The results of in vivo experiments indicated that the tumor growth of the 4T1 mouse tumor model was significantly inhibited. Additionally, toxicity studies showed that PDA@MB had good biocompatibility and safety. In conclusion, this study provides a promising chemo-photothermal therapy (CPT) nano-platform for precise and effective breast cancer therapy.
Collapse
Affiliation(s)
- Junke Leng
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Xiaofeng Dai
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Xiao Cheng
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Hao Zhou
- School of Food and Environment, Dalian University of Technology, Panjin, China
| | - Dong Wang
- Panjin People’s Hospital, Panjin, China
| | - Jing Zhao
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Kun Ma
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Changhao Cui
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Li Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Zhaoming Guo
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
- *Correspondence: Zhaoming Guo,
| |
Collapse
|
198
|
Park H, Otte A, Park K. Evolution of drug delivery systems: From 1950 to 2020 and beyond. J Control Release 2022; 342:53-65. [PMID: 34971694 PMCID: PMC8840987 DOI: 10.1016/j.jconrel.2021.12.030] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 12/21/2021] [Indexed: 02/03/2023]
Abstract
Modern drug delivery technology began in 1952 with the advent of the Spansule® sustained-release capsule technology, which can deliver a drug for 12 h after oral administration through an initial immediate dose followed by the remaining released gradually. Until the 1980s, oral and transdermal formulations providing therapeutic durations up to 24 h for small molecules dominated the drug delivery field and the market. The introduction of Lupron Depot® in 1989 opened the door for long-acting injectables and implantables, extending the drug delivery duration from days to months and occasionally years. Notably, the new technologies allowed long-term delivery of peptide and protein drugs, although limited to parenteral administration. The introduction of the first PEGylated protein, Adagen®, in 1990 marked the new era of PEGylation, resulting in Doxil® (doxorubicin in PEGylated liposome) in 1995, Movantik® (PEGylated naloxone - naloxegol) in 2014, and Onpattro® (Patisiran - siRNA in PEGylated lipid nanoparticle) in 2018. Drug-polymer complexes were introduced, e.g., InFed® (iron-dextran complex injection) in 1974 and Abraxane® (paclitaxel-albumin complex) in 2005. In 2000, both Mylotarg™ (antibody-drug conjugate - gemtuzumab ozogamicin) and Rapamune® (sirolimus nanocrystal formulation) were introduced. The year 2000 also marked the launching of the National Nanotechnology Initiative by the U.S. government, which was soon followed by the rest of the world. Extensive work on nanomedicine, particularly formulations designed to escape from endosomes after being taken by tumor cells, along with PEGylation technology, ultimately resulted in the timely development of lipid nanoparticle formulations for COVID-19 vaccine delivery in 2020. While the advances in drug delivery technologies for the last seven decades are breathtaking, they are only the tip of an iceberg of technologies that have yet to be utilized in an approved formulation or even to be discovered. As life expectancy continues to increase, more people require long-term care for various diseases. Filling the current and future unmet needs requires innovative drug delivery technologies to overcome age-old familiar hurdles, e.g., improving water-solubility of poorly soluble drugs, overcoming biological barriers, and developing more efficient long-acting depot formulations. The lessons learned from the past are essential assets for developing future drug delivery technologies implemented into products. As the development of COVID-19 vaccines demonstrated, meeting the unforeseen crisis of the uncertain future requires continuous cumulation of failures (as learning experiences), knowledge, and technologies. Conscious efforts of supporting diversified research topics in the drug delivery field are urgently needed more than ever.
Collapse
Affiliation(s)
- Haesun Park
- Akina, Inc., West Lafayette, IN 47906, United States of America
| | - Andrew Otte
- Purdue University, Departments of Biomedical Engineering and Pharmaceutics, West Lafayette, IN 47907, United States of America
| | - Kinam Park
- Akina, Inc., West Lafayette, IN 47906, United States of America; Purdue University, Departments of Biomedical Engineering and Pharmaceutics, West Lafayette, IN 47907, United States of America.
| |
Collapse
|
199
|
Discovery in polyethylene glycol immunogenicity: the characteristic of intergenerational inheritance of anti-polyethylene glycol IgG. Eur J Pharm Biopharm 2022; 172:89-100. [DOI: 10.1016/j.ejpb.2022.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 01/17/2022] [Accepted: 01/23/2022] [Indexed: 12/17/2022]
|
200
|
PEGylated nanoemulsions containing 1,2-distearoyl-sn-glycero-3-phosphoglycerol induced weakened accelerated blood clearance phenomenon. Drug Deliv Transl Res 2022; 12:2569-2579. [DOI: 10.1007/s13346-021-01111-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2021] [Indexed: 11/25/2022]
|