1
|
Münter R, Bak M, Thomsen ME, Parhamifar L, Stensballe A, Simonsen JB, Kristensen K, Andresen TL. Deciphering the monocyte-targeting mechanisms of PEGylated cationic liposomes by investigating the biomolecular corona. Int J Pharm 2024; 657:124129. [PMID: 38621615 DOI: 10.1016/j.ijpharm.2024.124129] [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: 01/02/2024] [Revised: 04/04/2024] [Accepted: 04/13/2024] [Indexed: 04/17/2024]
Abstract
Cationic liposomes specifically target monocytes in blood, rendering them promising drug-delivery tools for cancer immunotherapy, vaccines, and therapies for monocytic leukaemia. The mechanism behind this monocyte targeting ability is, however, not understood, but may involve plasma proteins adsorbed on the liposomal surfaces. To shed light on this, we investigated the biomolecular corona of three different types of PEGylated cationic liposomes, finding all of them to adsorb hyaluronan-associated proteins and proteoglycans upon incubation in human blood plasma. This prompted us to study the role of the TLR4 co-receptors CD44 and CD14, both involved in signalling and uptake pathways of proteoglycans and glycosaminoglycans. We found that separate inhibition of each of these receptors hampered the monocyte uptake of the liposomes in whole human blood. Based on clues from the biomolecular corona, we have thus identified two receptors involved in the targeting and uptake of cationic liposomes in monocytes, in turn suggesting that certain proteoglycans and glycosaminoglycans may serve as monocyte-targeting opsonins. This mechanistic knowledge may pave the way for rational design of future monocyte-targeting drug-delivery platforms.
Collapse
Affiliation(s)
- Rasmus Münter
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Martin Bak
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Mikkel E Thomsen
- Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark
| | - Ladan Parhamifar
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; Clinical Cancer Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Jens B Simonsen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Kasper Kristensen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Thomas L Andresen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
2
|
David CAW, de la Fonteyne-Blankestijn LJJ, Vermeulen JP, Plant-Hately AJ, Vandebriel RJ, Liptrott NJ. Application of KU812 cells for assessing complement activation related effects by nano(bio)materials. Biomed Pharmacother 2023; 163:114841. [PMID: 37167727 DOI: 10.1016/j.biopha.2023.114841] [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: 03/24/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/13/2023] Open
Abstract
Immunocompatibility issues related to nano(bio)materials, particularly liposomal formulations, involving activation of the complement system have been relatively well described however, they highlight the importance of preclinical evaluation of such interactions. These complement-mediated hypersensitivity reactions, in which basophils are implicated, are associated with complement activation-related pseudoallergy (CARPA). Ex vivo investigation of such events using primary basophils is technically challenging due to the relatively limited number of circulating basophils in peripheral blood. In the current work, the KU812 cell line has been applied as an in vitro model for basophil activation to investigate CARPA-related responses following exposure to test materials obtained from the REFINE consortium. To that end, we developed a standard operating procedure measuring a panel of cell-surface markers indicative of basophilic activation. Two laboratories performed the assays, demonstrating a clear difference in responses between liposomal and polymeric nano(bio)materials, while interlaboratory comparison of the standard operating procedure demonstrated reproducibility in results, between the two facilities. These results suggest the potential to use this protocol as a screening method for such responses however, validation using primary basophils is now warranted.
Collapse
Affiliation(s)
- Christopher A W David
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | | | - Jolanda P Vermeulen
- National Institute for Public Health & the Environment, Bilthoven, the Netherlands
| | - Alexander J Plant-Hately
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Rob J Vandebriel
- National Institute for Public Health & the Environment, Bilthoven, the Netherlands
| | - Neill J Liptrott
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
3
|
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: 5] [Impact Index Per Article: 2.5] [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
|
4
|
Platelet-Rich Plasma as an Alternative to Xenogeneic Sera in Cell-Based Therapies: A Need for Standardization. Int J Mol Sci 2022; 23:ijms23126552. [PMID: 35742995 PMCID: PMC9223511 DOI: 10.3390/ijms23126552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
There has been an explosion in scientific interest in using human-platelet-rich plasma (PRP) as a substitute of xenogeneic sera in cell-based therapies. However, there is a need to create standardization in this field. This systematic review is based on literature searches in PubMed and Web of Science databases until June 2021. Forty-one studies completed the selection criteria. The composition of PRP was completely reported in less than 30% of the studies. PRP has been used as PRP-derived supernatant or non-activated PRP. Two ranges could be identified for platelet concentration, the first between 0.14 × 106 and 0.80 × 106 platelets/µL and the second between 1.086 × 106 and 10 × 106 platelets/µL. Several studies have pooled PRP with a pool size varying from four to nine donors. The optimal dose for the PRP or PRP supernatant is 10%. PRP or PRP-derived supernatants a have positive effect on MSC colony number and size, cell proliferation, cell differentiation and genetic stability. The use of leukocyte-depleted PRP has been demonstrated to be a feasible alternative to xenogeneic sera. However, there is a need to improve the description of the PRP preparation methodology as well as its composition. Several items are identified and reported to create guidelines for future research.
Collapse
|
5
|
Ramos TI, Villacis-Aguirre CA, López-Aguilar KV, Santiago Padilla L, Altamirano C, Toledo JR, Santiago Vispo N. The Hitchhiker's Guide to Human Therapeutic Nanoparticle Development. Pharmaceutics 2022; 14:247. [PMID: 35213980 PMCID: PMC8879439 DOI: 10.3390/pharmaceutics14020247] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 02/06/2023] Open
Abstract
Nanomedicine plays an essential role in developing new therapies through novel drug delivery systems, diagnostic and imaging systems, vaccine development, antibacterial tools, and high-throughput screening. One of the most promising drug delivery systems are nanoparticles, which can be designed with various compositions, sizes, shapes, and surface modifications. These nanosystems have improved therapeutic profiles, increased bioavailability, and reduced the toxicity of the product they carry. However, the clinical translation of nanomedicines requires a thorough understanding of their properties to avoid problems with the most questioned aspect of nanosystems: safety. The particular physicochemical properties of nano-drugs lead to the need for additional safety, quality, and efficacy testing. Consequently, challenges arise during the physicochemical characterization, the production process, in vitro characterization, in vivo characterization, and the clinical stages of development of these biopharmaceuticals. The lack of a specific regulatory framework for nanoformulations has caused significant gaps in the requirements needed to be successful during their approval, especially with tests that demonstrate their safety and efficacy. Researchers face many difficulties in establishing evidence to extrapolate results from one level of development to another, for example, from an in vitro demonstration phase to an in vivo demonstration phase. Additional guidance is required to cover the particularities of this type of product, as some challenges in the regulatory framework do not allow for an accurate assessment of NPs with sufficient evidence of clinical success. This work aims to identify current regulatory issues during the implementation of nanoparticle assays and describe the major challenges that researchers have faced when exposing a new formulation. We further reflect on the current regulatory standards required for the approval of these biopharmaceuticals and the requirements demanded by the regulatory agencies. Our work will provide helpful information to improve the success of nanomedicines by compiling the challenges described in the literature that support the development of this novel encapsulation system. We propose a step-by-step approach through the different stages of the development of nanoformulations, from their design to the clinical stage, exemplifying the different challenges and the measures taken by the regulatory agencies to respond to these challenges.
Collapse
Affiliation(s)
- Thelvia I. Ramos
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, Concepción 4070386, Chile; (T.I.R.); (C.A.V.-A.)
- Grupo de Investigación en Sanidad Animal y Humana (GISAH), Carrera Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas—ESPE, Sangolquí 171103, Ecuador
| | - Carlos A. Villacis-Aguirre
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, Concepción 4070386, Chile; (T.I.R.); (C.A.V.-A.)
| | - Katherine V. López-Aguilar
- Carrera Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas—ESPE, Sangolquí 171103, Ecuador;
| | | | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Av. Brasil 2085, Valparaíso 2362803, Chile;
- Centro Regional de Estudios en Alimentos Saludables, Av. Universidad 330, Placilla, Sector Curauma, Valparaíso 2340000, Chile
| | - Jorge R. Toledo
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, Concepción 4070386, Chile; (T.I.R.); (C.A.V.-A.)
| | - Nelson Santiago Vispo
- School of Biological Sciences and Engineering, Yachay Tech University, Hda. San José s/n y Proyecto Yachay, Urcuquí 100119, Ecuador
| |
Collapse
|
6
|
Kong BS, Lee C, Cho YM. Protocol for the assessment of human T cell activation by real-time metabolic flux analysis. STAR Protoc 2022; 3:101084. [PMID: 35072113 PMCID: PMC8761778 DOI: 10.1016/j.xpro.2021.101084] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The elevation of glycolysis in autoreactive T cells is a key target for the prevention and treatment of T cell-related autoimmune diseases, such as type 1 diabetes (T1D). Here, we describe a simple and efficient protocol for isolating human peripheral blood mononuclear cells (PBMCs) and T cells, and the subsequent assessment of T cell glycolysis using Seahorse analyzer. This protocol is useful to analyze different subsets of T cells and applicable to different autoimmune disease models (i.e., T1D, multiple sclerosis). For complete details on the use and execution of this profile, please refer to Kong et al. (2021).
Collapse
Affiliation(s)
- Byung Soo Kong
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea,Corresponding author
| | - Changhan Lee
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA,Corresponding author
| | - Young Min Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea,Corresponding author
| |
Collapse
|
7
|
Shi D, Beasock D, Fessler A, Szebeni J, Ljubimova JY, Afonin KA, Dobrovolskaia MA. To PEGylate or not to PEGylate: Immunological properties of nanomedicine's most popular component, polyethylene glycol and its alternatives. Adv Drug Deliv Rev 2022; 180:114079. [PMID: 34902516 PMCID: PMC8899923 DOI: 10.1016/j.addr.2021.114079] [Citation(s) in RCA: 177] [Impact Index Per Article: 88.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 01/03/2023]
Abstract
Polyethylene glycol or PEG has a long history of use in medicine. Many conventional formulations utilize PEG as either an active ingredient or an excipient. PEG found its use in biotechnology therapeutics as a tool to slow down drug clearance and shield protein therapeutics from undesirable immunogenicity. Nanotechnology field applies PEG to create stealth drug carriers with prolonged circulation time and decreased recognition and clearance by the mononuclear phagocyte system (MPS). Most nanomedicines approved for clinical use and experimental nanotherapeutics contain PEG. Among the most recent successful examples are two mRNA-based COVID-19 vaccines that are delivered by PEGylated lipid nanoparticles. The breadth of PEG use in a wide variety of over the counter (OTC) medications as well as in drug products and vaccines stimulated research which uncovered that PEG is not as immunologically inert as it was initially expected. Herein, we review the current understanding of PEG's immunological properties and discuss them in the context of synthesis, biodistribution, safety, efficacy, and characterization of PEGylated nanomedicines. We also review the current knowledge about immunological compatibility of other polymers that are being actively investigated as PEG alternatives.
Collapse
Key Words
- Poly(ethylene)glycol, PEG, immunogenicity, immunology, nanomedicine, toxicity, anti-PEG antibodies, hypersensitivity, synthesis, drug delivery, biotherapeutics
Collapse
Affiliation(s)
- Da Shi
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Damian Beasock
- University of North Carolina Charlotte, Charlotte, NC, USA
| | - Adam Fessler
- University of North Carolina Charlotte, Charlotte, NC, USA
| | - Janos Szebeni
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary; SeroScience LCC, Budapest, Hungary; Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary
| | | | | | - Marina A Dobrovolskaia
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
8
|
Bhan A, Ansari K, Chen MY, Jandial R. Human induced pluripotent stem cell-derived platelets loaded with lapatinib effectively target HER2+ breast cancer metastasis to the brain. Sci Rep 2021; 11:16866. [PMID: 34654856 PMCID: PMC8521584 DOI: 10.1038/s41598-021-96351-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 08/06/2021] [Indexed: 01/09/2023] Open
Abstract
Prognosis of patients with HER2+ breast-to-brain-metastasis (BBM) is dismal even after current standard-of-care treatments, including surgical resection, whole-brain radiation, and systemic chemotherapy. Radiation and systemic chemotherapies can also induce cytotoxicity, leading to significant side effects. Studies indicate that donor-derived platelets can serve as immune-compatible drug carriers that interact with and deliver drugs to cancer cells with fewer side effects, making them a promising therapeutic option with enhanced antitumor activity. Moreover, human induced pluripotent stem cells (hiPSCs) provide a potentially renewable source of clinical-grade transfusable platelets that can be drug-loaded to complement the supply of donor-derived platelets. Here, we describe methods for ex vivo generation of megakaryocytes (MKs) and functional platelets from hiPSCs (hiPSC-platelets) in a scalable fashion. We then loaded hiPSC-platelets with lapatinib and infused them into BBM tumor-bearing NOD/SCID mouse models. Such treatment significantly increased intracellular lapatinib accumulation in BBMs in vivo, potentially via tumor cell-induced activation/aggregation. Lapatinib-loaded hiPSC-platelets exhibited normal morphology and function and released lapatinib pH-dependently. Importantly, lapatinib delivery to BBM cells via hiPSC-platelets inhibited tumor growth and prolonged survival of tumor-bearing mice. Overall, use of lapatinib-loaded hiPSC-platelets effectively reduced adverse effects of free lapatinib and enhanced its therapeutic efficacy, suggesting that they represent a novel means to deliver chemotherapeutic drugs as treatment for BBM.
Collapse
Affiliation(s)
- Arunoday Bhan
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA.
| | - Khairul Ansari
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
- Celcuity LLC, Minneapolis, MN, 55446, USA
| | - Mike Y Chen
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Rahul Jandial
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| |
Collapse
|
9
|
Kristensen K, Münter R, Kempen PJ, Thomsen ME, Stensballe A, Andresen TL. Isolation methods commonly used to study the liposomal protein corona suffer from contamination issues. Acta Biomater 2021; 130:460-472. [PMID: 34116227 DOI: 10.1016/j.actbio.2021.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022]
Abstract
The liposomal protein corona has been the focus of numerous studies, but there is still no consensus regarding its extent and composition. Rather, the literature is full of conflicting reports on the matter. To elucidate whether there could be a methodological explanation for this, we here scrutinize the efficiency of three commonly used liposome isolation methods at isolating stealth liposomes from human plasma. Firstly, we show that size-exclusion chromatography (SEC) in its standard form is prone to isolating unbound protein material together with the liposomes, but also that the method may be optimized to mitigate this issue. Secondly, we demonstrate that SEC in combination with membrane ultrafiltration is no better at removing the unbound protein material than SEC alone. Thirdly, we show that centrifugation is not able to pellet the liposomes. Overall, our results suggest that previous research on the liposomal protein corona may have suffered from significant methodological problems, in particular related to contaminant proteins interfering with the analysis of the protein corona. We believe that the data presented here may help guide future research around this challenge to reach a converging understanding about the properties of the protein corona on liposomes. STATEMENT OF SIGNIFICANCE: Upon administration into the circulatory system, liposomal drug carriers encounter an environment rich in proteins. These proteins may adsorb to the liposomes to form what is known as the protein corona, potentially governing the interactions of the liposomes with tissues and cells. However, despite decades of intense research efforts, there is currently no clear understanding about the extent and composition of the liposomal protein corona, making it impossible to assess its mechanistic importance. Here we report that the methods commonly used to isolate liposomes from blood plasma or serum to study the protein corona are susceptible to protein contamination. This may be the underlying technical reason for the current confusion about the characteristics of the liposomal protein corona.
Collapse
Affiliation(s)
- Kasper Kristensen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Rasmus Münter
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Paul J Kempen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Mikkel E Thomsen
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg Ø, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg Ø, Denmark
| | - Thomas L Andresen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
10
|
Kozma GT, Mészáros T, Bakos T, Hennies M, Bencze D, Uzonyi B, Győrffy B, Cedrone E, Dobrovolskaia MA, Józsi M, Szebeni J. Mini-Factor H Modulates Complement-Dependent IL-6 and IL-10 Release in an Immune Cell Culture (PBMC) Model: Potential Benefits Against Cytokine Storm. Front Immunol 2021; 12:642860. [PMID: 33995361 PMCID: PMC8113956 DOI: 10.3389/fimmu.2021.642860] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/24/2021] [Indexed: 12/15/2022] Open
Abstract
Cytokine storm (CS), an excessive release of proinflammatory cytokines upon overactivation of the innate immune system, came recently to the focus of interest because of its role in the life-threatening consequences of certain immune therapies and viral diseases, including CAR-T cell therapy and Covid-19. Because complement activation with subsequent anaphylatoxin release is in the core of innate immune stimulation, studying the relationship between complement activation and cytokine release in an in vitro CS model holds promise to better understand CS and identify new therapies against it. We used peripheral blood mononuclear cells (PBMCs) cultured in the presence of autologous serum to test the impact of complement activation and inhibition on cytokine release, testing the effects of liposomal amphotericin B (AmBisome), zymosan and bacterial lipopolysaccharide (LPS) as immune activators and heat inactivation of serum, EDTA and mini-factor H (mfH) as complement inhibitors. These activators induced significant rises of complement activation markers C3a, C4a, C5a, Ba, Bb, and sC5b-9 at 45 min of incubation, with or without ~5- to ~2,000-fold rises of IL-1α, IL-1β, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12, IL-13 and TNFα at 6 and 18 h later. Inhibition of complement activation by the mentioned three methods had differential inhibition, or even stimulation of certain cytokines, among which effects a limited suppressive effect of mfH on IL-6 secretion and significant stimulation of IL-10 implies anti-CS and anti-inflammatory impacts. These findings suggest the utility of the model for in vitro studies on CS, and the potential clinical use of mfH against CS.
Collapse
Affiliation(s)
- Gergely Tibor Kozma
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
| | - Tamás Mészáros
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
| | - Tamás Bakos
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | | | - Dániel Bencze
- MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Barbara Uzonyi
- MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Balázs Győrffy
- Second Department of Bioinformatics and Pediatrics, Semmelweis University, Budapest, Hungary
- Lendület Cancer Biomarker Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Edward Cedrone
- Nanotechnology Characterization Lab, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Marina A. Dobrovolskaia
- Nanotechnology Characterization Lab, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Mihály Józsi
- MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - János Szebeni
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
- Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary
| |
Collapse
|
11
|
Anitua E, Zalduendo M, Troya M, Alkhraisat MH. The influence of sodium citrate on the characteristics and biological activity of plasma rich in growth factors. Regen Med 2020; 15:2181-2192. [PMID: 33275449 DOI: 10.2217/rme-2020-0082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study investigated the effect of sodium citrate on the properties and biological activity of plasma rich in growth factors (PRGF). Methods: PRGF was obtained from trisodium citrate and plain extraction tubes. Hematological parameters, growth factors' release kinetics from both PRGF clots and their releasates' biological effect on human bone cells were evaluated. Results: The platelet enrichment factor, the growth factors' content and the release kinetic of PRGF were similar for both groups. The proliferation, collagen type I synthesis and tissue-nonspecific alkaline phosphatase activity of human osteoblasts showed no statistically significant differences. Conclusion: The use of sodium citrate does not influence the composition, the growth factors' release kinetics or the biological effect of PRGF, but it increases its clinical versatility.
Collapse
Affiliation(s)
| | | | - María Troya
- BTI - Biotechnology Institute, Vitoria, Spain
| | | |
Collapse
|
12
|
Ghosh S, Lalani R, Maiti K, Banerjee S, Patel V, Bhowmick S, Misra A. Optimization and efficacy study of synergistic vincristine coloaded liposomal doxorubicin against breast and lung cancer. Nanomedicine (Lond) 2020; 15:2585-2607. [DOI: 10.2217/nnm-2020-0169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aim: To improve the efficacy of poly-ethylene glycol (PEG)ylated liposomes coloaded with doxorubicin and vincristine against triple-negative breast cancer (TNBC) and non-small-cell lung cancer (NSCLC). Methods: The combinatorial index of the drugs was established using the Chou-Talalay method in MDA-MB-231 and A549 cell lines. The most effective ratio was co-encapsulated in factorial design optimized nanoliposomes which were characterized for similarity to clinical standard and evaluated in vitro and in vivo for therapeutic efficacy. Results & conclusion: The formulation exhibited more than 95% co-encapsulation, a size of 95.74 ± 2.65 nm and zeta potential of -9.17 ± 1.19 mV while having no significant differences in physicochemical and biochemical characteristics as compared with the clinical standard. Efficacy evaluation studies showed significantly improved cytotoxicity and tumor regression compared with liposomal doxorubicin indicating improvement in efficacy against TNBC and NSCLC.
Collapse
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
- Formulation Research & Development Department, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat-390012, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
| | - Kuntal Maiti
- Formulation Research & Development Department, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat-390012, India
| | - Shubhadeep Banerjee
- Formulation Research & Development Department, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat-390012, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
- Formulation Research & Development Department, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat-390012, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
- Pharmaceutical Research, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS University, VL Mehta Road, Vile Parle (W), Mumbai, Maharashtra - 400 056, Maharashtra India
| |
Collapse
|
13
|
Chen E, Chen BM, Su YC, Chang YC, Cheng TL, Barenholz Y, Roffler SR. Premature Drug Release from Polyethylene Glycol (PEG)-Coated Liposomal Doxorubicin via Formation of the Membrane Attack Complex. ACS NANO 2020; 14:7808-7822. [PMID: 32142248 DOI: 10.1021/acsnano.9b07218] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Anti-polyethylene glycol (PEG) antibodies are present in many healthy individuals as well as in patients receiving polyethylene glycol-functionalized drugs. Antibodies against PEG-coated nanocarriers can accelerate their clearance, but their impact on nanodrug properties including nanocarrier integrity is unclear. Here, we show that anti-PEG IgG and IgM antibodies bind to PEG molecules on the surface of PEG-coated liposomal doxorubicin (Doxil, Doxisome, LC-101, and Lipo-Dox), resulting in complement activation, formation of the membrane attack complex (C5b-9) in the liposomal membrane, and rapid release of encapsulated doxorubicin from the liposomes. Drug release depended on both classical and alternative pathways of complement activation. Doxorubicin release of up to 40% was also observed in rats treated with anti-PEG IgG and PEG-coated liposomal doxorubicin. Our results demonstrate that anti-PEG antibodies can disrupt the membrane integrity of PEG-coated liposomal doxorubicin through activation of complement, which may alter therapeutic efficacy and safety in patients with high levels of pre-existing antibodies against PEG.
Collapse
Affiliation(s)
- Even Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Cheng Su
- Department of Biological Sciences and Technology, National Chiao Tung University, Hsin-Chu 1001, Taiwan
| | - Yuan-Chih Chang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Tian-Lu Cheng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yechezekel Barenholz
- Department of Biochemistry, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
14
|
Mathews JA, Ni YG, Wang C, Peterson JE, Ray C, Zhao X, Duan D, Hamon S, Allinson J, Hokom M, Wegner G. Considerations for Soluble Protein Biomarker Blood Sample Matrix Selection. AAPS JOURNAL 2020; 22:38. [PMID: 31997095 DOI: 10.1208/s12248-020-0412-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/23/2019] [Indexed: 11/30/2022]
Abstract
Blood-based soluble protein biomarkers provide invaluable clinical information about patients and are used as diagnostic, prognostic, and pharmacodynamic markers. The most commonly used blood sample matrices are serum and different types of plasma. In drug development research, the impact of sample matrix selection on successful protein biomarker quantification is sometimes overlooked. The sample matrix for a specific analyte is often chosen based on prior experience or literature searches, without good understanding of the possible effects on analyte quantification. Using a data set of 32 different soluble protein markers measured in matched serum and plasma samples, we examined the differences between serum and plasma and discussed how platelet or immune cell activation can change the quantified concentration of the analyte. We have also reviewed the effect of anticoagulant on analyte quantification. Finally, we provide specific recommendations for biomarker sample matrix selection and propose a systematic and data-driven approach for sample matrix selection. This review is intended to raise awareness of the impact and considerations of sample matrix selection on biomarker quantification.
Collapse
Affiliation(s)
- Joel A Mathews
- Ionis Pharmaceuticals, 2855 Gazelle Rd., Carlsbad, California, 92010, USA.
| | - Yan G Ni
- Regeneron, Tarrytown, New York, USA
| | | | | | - Chad Ray
- Zoetis, Kalamazoo, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Őrfi E, Mészáros T, Hennies M, Fülöp T, Dézsi L, Nardocci A, Rosivall L, Hamar P, Neun BW, Dobrovolskaia MA, Szebeni J, Szénási G. Acute physiological changes caused by complement activators and amphotericin B-containing liposomes in mice. Int J Nanomedicine 2019; 14:1563-1573. [PMID: 30880965 PMCID: PMC6396670 DOI: 10.2147/ijn.s187139] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Undesirable complement (C) activation by nanomedicines can entail an adverse immune reaction known as C activation-related pseudoallergy (CARPA) in sensitive patients. The syndrome includes cardiopulmonary, hemodynamic, and a variety of other physiological changes that have been well described in man, pigs, dogs, and rats. However, the information on CARPA is scarce and ambiguous in mice, a species widely used in preclinical studies. The present study aimed to fill this gap by exploring signs of CARPA in mice following i.v. administration of AmBisome and Abelcet, which are nano-formulations of Amphotericin B with high risk to cause CARPA. MATERIALS AND METHODS Anesthetized NMRI mice were intravenously injected with liposomal amphotericin B (Abelcet and AmBisome; 30-300 mg phospholipid/kg), drug-free high cholesterol multilamellar vesicles (HC-MLV), and positive controls, cobra venom factor (CVF) and zymosan, followed by the measurement of blood pressure (BP), heart rate, white blood cell, and platelet counts and plasma thromboxane B2 (TXB2) levels. C activation was assessed by C3a ELISA, a C3 consumption assay (PAN-C3) and a modified sheep red blood cell hemolytic assay. RESULTS All test agents, except HC-MLV, caused transient hypertension, thrombocytopenia, and elevation of plasma TXB2, which were paralleled by significant rises of plasma C3a in CVF and zymosan-treated animals, wherein the initial hypertension turned into hypotension and shock. Abelcet and AmBisome caused minor, delayed rise of C3a that was not associated with hypertension. The C3a receptor inhibitor SB-290157 attenuated the hypertension caused by Abelcet and decreased the BP thereafter. CONCLUSION The parallelism between C3a anaphylatoxin production and severity of physiological changes caused by the different agents is consistent with CARPA underlying these changes. Although the reactive dose of liposomal phospholipids was substantially higher than that in other species (pigs, dogs), the mouse seems suitable for studying the mechanism of hypersensitivity reactions to liposomal formulations of amphotericin B, a frequent side effect of these drugs.
Collapse
Affiliation(s)
- Erik Őrfi
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary,
- SeroScience LCC., Cambridge, MA, USA,
| | - Tamás Mészáros
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary,
- SeroScience LCC., Cambridge, MA, USA,
| | | | - Tamás Fülöp
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary,
- SeroScience LCC., Cambridge, MA, USA,
| | - László Dézsi
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary,
- SeroScience LCC., Cambridge, MA, USA,
| | - Alexander Nardocci
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary,
| | - László Rosivall
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary,
- SeroScience LCC., Cambridge, MA, USA,
| | - Péter Hamar
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Barry W Neun
- Nanotechnology Characterization Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - János Szebeni
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary,
- SeroScience LCC., Cambridge, MA, USA,
- Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary,
| | - Gábor Szénási
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary,
- Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
16
|
Dobrovolskaia M, Neun BW, Szénási G, Szebeni J. Plasma samples from mouse strains and humans demonstrate different susceptibilities to complement activation. PRECISION NANOMEDICINE 2018. [DOI: 10.33218/prnano1(3).181029.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Complement activation can be evaluated in vitro using plasma or serum from animals and human donors, and in vivo using animal models. Despite many years of research, there is no harmonized approach for the selection of matrix and animal models. Herein, we present an in vitro study investigating intra- and inter-species variability in the complement activation. We used the liposomal formulation of amphotericin, Ambisome, as a model particle to assess the magnitude of the complement activation in plasma derived from various mouse strains and individual human donors. We demonstrated that mouse strains differ in the magnitude of the complement activation by liposomes and cobra venom factor (CVF) in vitro. Inter-individual variability in complement activation by Ambisome and CVF was also observed when plasma from individual human donors was analyzed. Such variability in both mouse and human plasma could not be explained by the levels of complement regulatory factors H and I. Moreover, even though mouse plasma was less sensitive to the complement activation by CVF than human plasma, it was equally sensitive to the activation by Ambisome. Our study demonstrates the importance of mouse strain selection for in vitro complement activation analysis. It also shows that traditional positive controls (e.g., CVF) are not predictive of the degree of complement activation by nanomedicines. The study also suggests that besides complement inhibitory factors, other elements contribute to the inter- and intra-species variability in complement activation by nanomedicines.
Collapse
|
17
|
Mészáros T, Kozma GT, Shimizu T, Miyahara K, Turjeman K, Ishida T, Barenholz Y, Urbanics R, Szebeni J. Involvement of complement activation in the pulmonary vasoactivity of polystyrene nanoparticles in pigs: unique surface properties underlying alternative pathway activation and instant opsonization. Int J Nanomedicine 2018; 13:6345-6357. [PMID: 30349254 PMCID: PMC6187999 DOI: 10.2147/ijn.s161369] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background It has been proposed that many hypersensitivity reactions to nanopharmaceuticals represent complement (C)-activation-related pseudoallergy (CARPA), and that pigs provide a sensitive animal model to study the phenomenon. However, a recent study suggested that pulmonary hypertension, the pivotal symptom of porcine CARPA, is not mediated by C in cases of polystyrene nanoparticle (PS-NP)-induced reactions. Goals To characterize PS-NPs and reexamine the contribution of CARPA to their pulmonary reactivity in pigs. Study design C activation by 200, 500, and 750 nm (diameter) PS-NPs and their opsonization were measured in human and pig sera, respectively, and correlated with hemodynamic effects of the same NPs in pigs in vivo. Methods Physicochemical characterization of PS-NPs included size, ζ-potential, cryo-transmission electron microscopy, and hydrophobicity analyses. C activation in human serum was measured by ELISA and opsonization of PS-NPs in pig serum by Western blot and flow cytometry. Pulmonary vasoactivity of PS-NPs was quantified in the porcine CARPA model. Results PS-NPs are monodisperse, highly hydrophobic spheres with strong negative surface charge. In human serum, they caused size-dependent, significant rises in C3a, Bb, and sC5b-9, but not C4d. Exposure to pig serum led within minutes to deposition of C5b-9 and opsonic iC3b on the NPs, and opsonic iC3b fragments (C3dg, C3d) also appeared in serum. PS-NPs caused major hemodynamic changes in pigs, primarily pulmonary hypertension, on the same time scale (minutes) as iC3b fragmentation and opsonization proceeded. There was significant correlation between C activation by different PS-NPs in human serum and pulmonary hypertension in pigs. Conclusion PS-NPs have extreme surface properties with no relevance to clinically used nanomedicines. They can activate C via the alternative pathway, entailing instantaneous opsonization of NPs in pig serum. Therefore, rather than being solely C-independent reactivity, the mechanism of PS-NP-induced hypersensitivity in pigs may involve C activation. These data are consistent with the “double-hit” concept of nanoparticle-induced hypersensitivity reactions involving both CARPA and C-independent pseudoallergy.
Collapse
Affiliation(s)
- Tamás Mészáros
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, Budapest, Hungary, .,SeroScience Ltd, Budapest, Hungary,
| | | | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Koga Miyahara
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Keren Turjeman
- Laboratory of Membrane and Liposome Research, Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Rudolf Urbanics
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, Budapest, Hungary, .,SeroScience Ltd, Budapest, Hungary,
| | - János Szebeni
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, Budapest, Hungary, .,SeroScience Ltd, Budapest, Hungary, .,Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary,
| |
Collapse
|
18
|
Understanding the Role of Anti-PEG Antibodies in the Complement Activation by Doxil in Vitro. Molecules 2018; 23:molecules23071700. [PMID: 30002298 PMCID: PMC6100003 DOI: 10.3390/molecules23071700] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 11/27/2022] Open
Abstract
Infusion reactions (IRs) are common immune-mediated side effects in patients treated with a variety of drug products, including, but not limited to, nanotechnology formulations. The mechanism of IRs is not fully understood. One of the best studied mechanisms of IRs to nanomedicines is the complement activation. However, it is largely unknown why some patients develop reactions to nanomedicines while others do not, and why some nanoparticles are more reactogenic than others. One of the theories is that the pre-existing anti-polyethylene glycol (PEG) antibodies initiate the complement activation and IRs in patients. In this study, we investigated this hypothesis in the case of PEGylated liposomal doxorubicin (Doxil), which, when used in a clinical setting, is known to induce IRs; referred to as complement activation-related pseudoallergy (CARPA) in sensitive individuals. We conducted the study in vitro using plasma derived from C57BL/6 mice and twenty human donor volunteers. We used mouse plasma to test a library of well-characterized mouse monoclonal antibodies with different specificity and affinity to PEG as it relates to the complement activation by Doxil. We determined the levels of pre-existing polyclonal antibodies that bind to PEG, methoxy-PEG, and PEGylated liposomes in human plasma, and we also assessed complement activation by Doxil and concentrations of complement inhibitory factors H and I in these human plasma specimens. The affinity, specificity, and other characteristics of the human polyclonal antibodies are not known at this time. Our data demonstrate that under in vitro conditions, some anti-PEG antibodies contribute to the complement activation by Doxil. Such contribution, however, needs to be considered in the context of other factors, including, but not limited to, antibody class, type, clonality, epitope specificity, affinity, and titer. In addition, our data contribute to the knowledge base used to understand and improve nanomedicine safety.
Collapse
|