1
|
Jahromi BR, Zamotin V, Code C, Netti E, Lorey MB, Alitalo K, Öörni K, Laakso A, Tulamo R, Niemelä M. Immunoliposomes for detection of rupture-prone intracranial aneurysms. Acta Neurochir (Wien) 2023; 165:3353-3360. [PMID: 37749289 PMCID: PMC10624708 DOI: 10.1007/s00701-023-05770-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/15/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND It is estimated that significant (3.2%) of population carries intracranial aneurysm (IA). An increasing number of imaging studies have caused that the chance of finding an incidental aneurysm is becoming more common. Since IA rupture causes subarachnoidal hemorrhage (SAH) and have significant mortality and morbidity prophylactic treatment should be considered when IA is detected. The benefit and risk of treatment of IA is based on epidemiological estimate which takes account patient and aneurysm characteristics. However we know that aneurysm rupture is biological process where inflammation of aneurysm wall is actively leading to degeneration of aneurysm wall and finally weakens it until it bursts. Until now, there have not been imaging method to detect inflammatory process of aneurysm wall METHODS: We created targeting immunoliposome for use in the imaging of aneurysm. Immunoliposome comprises antibodies against at least one vascular inflammatory marker associated with aneurysm inflammation and a label and/or a contrast agent. RESULTS Histological analysis of IAs where immunoliposome comprises antibodies against vascular inflammation with a label shows promising results for selectively detecting aneurysms inflammation. In magnetic resonance imaging (MRI) we were able to detect immunoliposomes carrying gadolinium. CONCLUSION Our work opens a new avenue for using contrast labeled immunoliposomes for detecting rupture-prone aneurysms. Immunoliposomes can cary gadolinium and selectively bind to inflammatory section of aneurysm that can be detected with MRI. Further research is needed to develop immunoliposomes to be used with MRI in humans to target treatment to those patients who benefit from it the most.
Collapse
Affiliation(s)
- Behnam Rezai Jahromi
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Topeliuksenkatu 5, 00260, Helsinki, Finland.
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland.
| | - Vladimir Zamotin
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
| | - Christian Code
- PHYLIFE: Physical Life Science, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Odense, Denmark
| | - Eliisa Netti
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Topeliuksenkatu 5, 00260, Helsinki, Finland
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
| | - Martina B Lorey
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, Finland
| | - Kari Alitalo
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, Finland
| | - Katariina Öörni
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, Finland
| | - Aki Laakso
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Topeliuksenkatu 5, 00260, Helsinki, Finland
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
| | - Riikka Tulamo
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
- Department of Vascular Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mika Niemelä
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Topeliuksenkatu 5, 00260, Helsinki, Finland
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
| |
Collapse
|
2
|
Rezazadeh L, Pourmoradian S, Tutunchi H, Farrin N, Radkhah N, Ostadrahimi A. The effects of probiotics on VCAM-1 and ICAM-1: A systematic review and meta-analysis of randomized controlled trials. Clin Nutr ESPEN 2023; 54:60-67. [PMID: 36963899 DOI: 10.1016/j.clnesp.2023.01.009] [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: 10/26/2022] [Revised: 12/17/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023]
Abstract
BACKGROUND We performed the present systematic review and meta-analysis of randomized controlled trials (RCTs) to evaluate the effects of probiotics on intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) levels in adults. METHODS A systematic search current to April 2022 was performed in MEDLINE, EMBASE, and Cochrane Database using relevant keywords to detect eligible articles. A random-effects model was used to estimate the standardized mean difference (SMD) and 95% confidence interval (95% CI). RESULTS Six eligible trials were included in the final analysis. The pooled analysis revealed that there was a significant reduction in VCAM-1 from baseline to post-probiotic course with standardized mean difference [SMD: -0.66 ng/ml; 95% CI: -1.09, -0.23 ng/ml; P = 0.003]. The effects of probiotic intake on VCAM-1 were more pronounced when it was received via supplements [SMD: -0.61 ng/ml; 95% CI: -1.08, -0.14 ng/ml; P = 0.010], for 12 weeks [SMD: -0.60 ng/ml; 95% CI: -1.09, -0.12 ng/ml; P = 0.014] and when it was prescribed for individuals with metabolic syndrome [SMD: -0.79 ng/ml; 95% CI: -1.40, -0.19 ng/ml; P = 0.010]. Moreover, VCAM-1 levels were decreased in the subgroup of multispecies probiotic regiments [SMD: -0.71 ng/ml; 95% CI: -1.38, -0.04 ng/ml; P = 0.039]. CONCLUSION Our study demonstrates potential beneficial effects of probiotics on VCAM-1 in adults. However, more larger-scale, long-time RCTs are needed to confirm the accurate effect of probiotics on endothelial dysfunction biomarkers.
Collapse
Affiliation(s)
- Leila Rezazadeh
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Pourmoradian
- Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Helda Tutunchi
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Farrin
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Radkhah
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
YÜKSEL A, DAĞLIOĞLU Y. Kanser Tedavisi İçin MikroRNA’ların Çok İşlevli Nano-taşıyıcılar İle Dağıtımı. ARŞIV KAYNAK TARAMA DERGISI 2023. [DOI: 10.17827/aktd.1181394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Hücre proliferasyonu ve apoptozis gibi kanserden sorumlu biyolojik süreçlerde etkili olan miRNA’lar, farklı kanser türleri ve evrelerinin teşhis ve tedavisinde yeni biyobelirteçler olarak işlev görür. Bunun yanı sıra bazı miRNA’ların onkogen ve tümör baskılayıcı özelliği nanoteknoloji ile entegre edilmesiyle kanser oluşumunu engeller. Son yıllarda miRNA’ların kanser tedavisinde kullanılmasını sağlayan diğer bir yaklaşım ise nano-taşıyıcılardır. İlaçlar, peptitler veya genler gibi aktif bileşikleri taşımak için geliştirilen bu nano-taşıyıcıların kanser tedavisinde kullanımları umut vadetmektedir. Bu derleme, miRNA dağıtımında kullanılan nano-taşıyıcı türleri hakkında kısa bir bilgi sunmaktadır. Ayrıca nanoteknolojideki gelişmelerle birlikte miRNA’ların kanser teşhis ve tedavisinde kullanımın yanısıra gen susturma mekanizması olan RNA interferansından kısaca bahsedilmektedir.
Collapse
|
4
|
van Kerkhof P, Kralj T, Spanevello F, van Bloois L, Jordens I, van der Vaart J, Jamieson C, Merenda A, Mastrobattista E, Maurice MM. RSPO3 Furin domain-conjugated liposomes for selective drug delivery to LGR5-high cells. J Control Release 2023; 356:72-83. [PMID: 36813038 DOI: 10.1016/j.jconrel.2023.02.025] [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: 09/02/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
The transmembrane receptor LGR5 potentiates Wnt/β-catenin signaling by binding both secreted R-spondin (RSPOs) and the Wnt tumor suppressors RNF43/ZNRF3, directing clearance of RNF43/ZNRF3 from the cell surface. Besides being widely used as a stem cell marker in various tissues, LGR5 is overexpressed in many types of malignancies, including colorectal cancer. Its expression characterizes a subpopulation of cancer cells that play a crucial role in tumor initiation, progression and cancer relapse, known as cancer stem cells (CSCs). For this reason, ongoing efforts are aimed at eradicating LGR5-positive CSCs. Here, we engineered liposomes decorated with different RSPO proteins to specifically detect and target LGR5-positive cells. Using fluorescence-loaded liposomes, we show that conjugation of full-length RSPO1 to the liposomal surface mediates aspecific, LGR5-independent cellular uptake, largely mediated by heparan sulfate proteoglycan binding. By contrast, liposomes decorated only with the Furin (FuFu) domains of RSPO3 are taken up by cells in a highly specific, LGR5-dependent manner. Moreover, encapsulating doxorubicin in FuFuRSPO3 liposomes allowed us to selectively inhibit the growth of LGR5-high cells. Thus, FuFuRSPO3-coated liposomes allow for the selective detection and ablation of LGR5-high cells, providing a potential drug delivery system for LGR5-targeted anti-cancer strategies.
Collapse
Affiliation(s)
- Peter van Kerkhof
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Tomica Kralj
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Francesca Spanevello
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Louis van Bloois
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Ingrid Jordens
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Jelte van der Vaart
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Cara Jamieson
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Alessandra Merenda
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Enrico Mastrobattista
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands.
| | - Madelon M Maurice
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
5
|
Weichwald C, Zettl I, Ellinger I, Niespodziana K, Waltl EE, Villazala-Merino S, Ivanov D, Eckl-Dorna J, Niederberger-Leppin V, Valenta R, Flicker S. Antibody Conjugates Bispecific for Pollen Allergens and ICAM-1 with Potential to Prevent Epithelial Allergen Transmigration and Rhinovirus Infection. Int J Mol Sci 2023; 24:ijms24032725. [PMID: 36769047 PMCID: PMC9917280 DOI: 10.3390/ijms24032725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Allergy and rhinovirus (RV) infections are major triggers for rhinitis and asthma, causing a socioeconomic burden. As RVs and allergens may act synergistically to promote airway inflammation, simultaneous treatment strategies for both causative agents would be innovative. We have previously identified the transmembrane glycoprotein intercellular adhesion molecule 1 (ICAM-1) as an anchor for antibody conjugates bispecific for ICAM-1 and Phleum pratense (Phl p) 2, a major grass pollen allergen, to block allergen transmigration through the epithelial barrier. Since ICAM-1 is a receptor for the major group RVs, we speculated that our bispecific antibody conjugates may protect against RV infection. Therefore, we created antibody conjugates bispecific for ICAM-1 and the major grass pollen allergen Phl p 5 and analyzed their capacity to affect allergen penetration and RV infection. Bispecific antibody conjugates significantly reduced the trans-epithelial migration of Phl p 5 and thus the basolateral Phl p 5 concentration and allergenic activity as determined by humanized rat basophilic leukemia cells and inhibited RV infection of cultured epithelial cells. A reduction in allergenic activity was obtained only through the prevention of allergen transmigration because the Phl p 5-specific IgG antibody did not block the allergen-IgE interaction. Our results indicate the potential of allergen/ICAM-1-specific antibody conjugates as a topical treatment strategy for allergy and RV infections.
Collapse
Affiliation(s)
- Christina Weichwald
- Division of Immunopathology, Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Ines Zettl
- Division of Immunopathology, Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Isabella Ellinger
- Division of Cellular and Molecular Pathophysiology, Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Katarzyna Niespodziana
- Division of Immunopathology, Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Eva E. Waltl
- Department of Otorhinolaryngology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Daniel Ivanov
- Division of Immunopathology, Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Rudolf Valenta
- Division of Immunopathology, Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
- National Research Centre (NRC) Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, 115478 Moscow, Russia
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Sabine Flicker
- Division of Immunopathology, Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: ; Tel.: +43-1-40400-51150
| |
Collapse
|
6
|
Zettl I, Ivanova T, Zghaebi M, Rutovskaya MV, Ellinger I, Goryainova O, Kollárová J, Villazala-Merino S, Lupinek C, Weichwald C, Drescher A, Eckl-Dorna J, Tillib SV, Flicker S. Generation of high affinity ICAM-1-specific nanobodies and evaluation of their suitability for allergy treatment. Front Immunol 2022; 13:1022418. [DOI: 10.3389/fimmu.2022.1022418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
The nasal cavity is an important site of allergen entry. Hence, it represents an organ where trans-epithelial allergen penetration and subsequent IgE-mediated allergic inflammation can potentially be inhibited. Intercellular adhesion molecule 1 (ICAM-1) is highly expressed on the surface of respiratory epithelial cells in allergic patients. It was identified as a promising target to immobilize antibody conjugates bispecific for ICAM-1 and allergens and thereby block allergen entry. We have previously characterized a nanobody specific for the major birch pollen allergen Bet v 1 and here we report the generation and characterization of ICAM-1-specific nanobodies. Nanobodies were obtained from a camel immunized with ICAM-1 and a high affinity binder was selected after phage display (Nb44). Nb44 was expressed as recombinant protein containing HA- and His-tags in Escherichia coli (E.coli) and purified via affinity chromatography. SDS-PAGE and Western blot revealed a single band at approximately 20 kDa. Nb44 bound to recombinant ICAM-1 in ELISA, and to ICAM-1 expressed on the human bronchial epithelial cell line 16HBE14o- as determined by flow cytometry. Experiments conducted at 4°C and at 37°C, to mimic physiological conditions, yielded similar percentages (97.2 ± 1.2% and 96.7 ± 1.5% out of total live cells). To confirm and visualize binding, we performed immunofluorescence microscopy. While Texas Red Dextran was rapidly internalized Nb44 remained localized on the cell surface. Additionally, we determined the strength of Nb44 and ICAM-1 interaction using surface plasmon resonance (SPR). Nb44 bound ICAM-1 with high affinity (10-10 M) and had slow off-rates (10-4 s-1). In conclusion, our results showed that the selected ICAM-1-specific nanobody bound ICAM-1 with high affinity and was not internalized. Thus, it could be further used to engineer heterodimers with allergen-specific nanobodies in order to develop topical treatments of pollen allergy.
Collapse
|
7
|
Singh M, Thakur M, Mishra M, Yadav M, Vibhuti R, Menon AM, Nagda G, Dwivedi VP, Dakal TC, Yadav V. Gene regulation of intracellular adhesion molecule-1 (ICAM-1): A molecule with multiple functions. Immunol Lett 2021; 240:123-136. [PMID: 34715236 DOI: 10.1016/j.imlet.2021.10.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 01/04/2023]
Abstract
Intracellular adhesion molecule 1 (ICAM-1) is one of the most extensively studied inducible cell adhesion molecules which is responsible for several immune functions like T cell activation, extravasation, inflammation, etc. The molecule is constitutively expressed over the cell surface and is regulated up / down in response to inflammatory mediators like cellular stress, proinflammatory cytokines, viral infection. These stimuli modulate the expression of ICAM-1 primarily through regulating the ICAM-1 gene transcription. On account of the presence of various binding sites for NF-κB, AP-1, SP-1, and many other transcription factors, the architecture of the ICAM-1 promoter become complex. Transcription factors in union with other transcription factors, coactivators, and suppressors promote their assembly in a stereospecific manner on ICAM-1 promoter which mediates ICAM-1 regulation in response to different stimuli. Along with transcriptional regulation, epigenetic modifications also play a pivotal role in controlling ICAM-1 expression on different cell types. In this review, we summarize the regulation of ICAM-1 expression both at the transcriptional as well as post-transcriptional level with an emphasis on transcription factors and signaling pathways involved.
Collapse
Affiliation(s)
- Mona Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067 India
| | - Mony Thakur
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| | - Manish Mishra
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research- Institute of Microbial Technology, Chandigarh-160036 India
| | - Manisha Yadav
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research- Institute of Microbial Technology, Chandigarh-160036 India
| | - Rajkamal Vibhuti
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| | - Athira M Menon
- Genome and computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001 India
| | - Girima Nagda
- Department of Zoology, Mohanlal Sukhadia University, Udaipur, Rajasthan-313001 India
| | - Ved Prakash Dwivedi
- International Centre for Genetic Engineering and Biotechnology, ICGEB Campus, Aruna Asaf Ali Marg, New Delhi-110067 India
| | - Tikam Chand Dakal
- Genome and computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001 India
| | - Vinod Yadav
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| |
Collapse
|
8
|
Myeloperoxidase: A versatile mediator of endothelial dysfunction and therapeutic target during cardiovascular disease. Pharmacol Ther 2020; 221:107711. [PMID: 33137376 DOI: 10.1016/j.pharmthera.2020.107711] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Myeloperoxidase (MPO) is a prominent mammalian heme peroxidase and a fundamental component of the innate immune response against microbial pathogens. In recent times, MPO has received considerable attention as a key oxidative enzyme capable of impairing the bioactivity of nitric oxide (NO) and promoting endothelial dysfunction; a clinically relevant event that manifests throughout the development of inflammatory cardiovascular disease. Increasing evidence indicates that during cardiovascular disease, MPO is released intravascularly by activated leukocytes resulting in its transport and sequestration within the vascular endothelium. At this site, MPO catalyzes various oxidative reactions that are capable of promoting vascular inflammation and impairing NO bioactivity and endothelial function. In particular, MPO catalyzes the production of the potent oxidant hypochlorous acid (HOCl) and the catalytic consumption of NO via the enzyme's NO oxidase activity. An emerging paradigm is the ability of MPO to also influence endothelial function via non-catalytic, cytokine-like activities. In this review article we discuss the implications of our increasing knowledge of the versatility of MPO's actions as a mediator of cardiovascular disease and endothelial dysfunction for the development of new pharmacological agents capable of effectively combating MPO's pathogenic activities. More specifically, we will (i) discuss the various transport mechanisms by which MPO accumulates into the endothelium of inflamed or diseased arteries, (ii) detail the clinical and basic scientific evidence identifying MPO as a significant cause of endothelial dysfunction and cardiovascular disease, (iii) provide an up-to-date coverage on the different oxidative mechanisms by which MPO can impair endothelial function during cardiovascular disease including an evaluation of the contributions of MPO-catalyzed HOCl production and NO oxidation, and (iv) outline the novel non-enzymatic mechanisms of MPO and their potential contribution to endothelial dysfunction. Finally, we deliver a detailed appraisal of the different pharmacological strategies available for targeting the catalytic and non-catalytic modes-of-action of MPO in order to protect against endothelial dysfunction in cardiovascular disease.
Collapse
|
9
|
Kuhn J, Smirnov A, Criss AK, Columbus L. Quantifying Carcinoembryonic Antigen-like Cell Adhesion Molecule-Targeted Liposome Delivery Using Imaging Flow Cytometry. Mol Pharm 2019; 16:2354-2363. [PMID: 30995063 DOI: 10.1021/acs.molpharmaceut.8b01274] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Carcinoembryonic antigen-like cell adhesion molecules (CEACAMs) are human cell-surface proteins that can exhibit increased expression on tumor cells and are thus a potential target for novel tumor-seeking therapeutic delivery methods. We hypothesize that engineered nanoparticles containing a known interaction partner of CEACAM, Neisseria gonorrhoeae outer membrane protein Opa, can be used to deliver cargo to specific cellular targets. In this study, the cell association and uptake of protein-free liposomes and Opa proteoliposomes into CEACAM-expressing cells were measured using imaging flow cytometry. A size-dependent internalization of liposomes into HeLa cells was observed through endocytic pathways. Opa-dependent, CEACAM1-mediated uptake of liposomes into HeLa cells was observed, with limited colocalization with endosomal and lysosomal trafficking compartments. Given the overexpression of CEACAM1 on several distinct cancers and interest in using CEACAM1 as a component in treatment strategies, these results support further pursuit of investigating Opa-dependent specificity and the internalization mechanism for therapeutic delivery.
Collapse
|
10
|
Chirio D, Peira E, Sapino S, Dianzani C, Barge A, Muntoni E, Morel S, Gallarate M. Stearoyl-Chitosan Coated Nanoparticles Obtained by Microemulsion Cold Dilution Technique. Int J Mol Sci 2018; 19:ijms19123833. [PMID: 30513699 PMCID: PMC6321505 DOI: 10.3390/ijms19123833] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/23/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022] Open
Abstract
Chitosan is an excipient which has been studied thoroughly in research works thanks to its positive characteristics such as muco-adhesiveness and ability to open epithelial-tight-junctions. In this article, lipophilic stearoyl chitosan (ST-CS) was synthetized in order to anchor this polymer to lipid nanoparticles and prepare ST-CS-coated nanoparticles (ST-CS-NP) using the microemulsion cold dilution technique. Curcumin (CURC) was used as model drug. CURC-ST-CS-NP were characterized by dimensional analysis, zeta potential, drug entrapment, drug release; tested in vitro on Human Umbilical Vein Endothelial Cell (HUVEC) cells to study its cytotoxicity and on human pancreatic cancer cells (PANC-1) to determine inhibition ability; tested in rats to determine CURC blood profiles and biodistribution. CURC-ST-CS-NP had mean diameters in the range 200–400 nm and CURC entrapment up to 73%. These systems did not show cytotoxicity on HUVEC cells at all tested dilutions and revealed to be more effective than free CURC solution on PANC-1 cells at 5 and 10 µM CURC. Blood profile studies evidenced as CURC entrapment in NP prolonged the permanence of drug in the systemic circulation compared to CURC solution due to a certain stealth property of NP, probably attributable to hydrophilic chitosan coating. Biodistribution studies showed a smaller CURC concentration in RES organs when CURC-ST-CS-NP were administered.
Collapse
Affiliation(s)
- Daniela Chirio
- University of Turin, Dipartimento di Scienza e Tecnologia del Farmaco, via Giuria 9, Torino 10125, Italy.
| | - Elena Peira
- University of Turin, Dipartimento di Scienza e Tecnologia del Farmaco, via Giuria 9, Torino 10125, Italy.
| | - Simona Sapino
- University of Turin, Dipartimento di Scienza e Tecnologia del Farmaco, via Giuria 9, Torino 10125, Italy.
| | - Chiara Dianzani
- University of Turin, Dipartimento di Scienza e Tecnologia del Farmaco, via Giuria 9, Torino 10125, Italy.
| | - Alessandro Barge
- University of Turin, Dipartimento di Scienza e Tecnologia del Farmaco, via Giuria 9, Torino 10125, Italy.
| | - Elisabetta Muntoni
- University of Turin, Dipartimento di Scienza e Tecnologia del Farmaco, via Giuria 9, Torino 10125, Italy.
| | - Silvia Morel
- Amedeo Avogadro University of Eastern Piedmont, Dipartimento di Scienze del Farmaco, Largo Donegani 2/3, Novara 28100, Italy.
| | - Marina Gallarate
- University of Turin, Dipartimento di Scienza e Tecnologia del Farmaco, via Giuria 9, Torino 10125, Italy.
| |
Collapse
|
11
|
Merino M, Zalba S, Garrido MJ. Immunoliposomes in clinical oncology: State of the art and future perspectives. J Control Release 2018; 275:162-176. [DOI: 10.1016/j.jconrel.2018.02.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/09/2018] [Accepted: 02/10/2018] [Indexed: 02/02/2023]
|
12
|
Abstract
In this study, fast and slow nitric oxide (NO)-releasing liposomes (half-lives of 2.5 and >72 h, respectively) were prepared by encapsulation of N-propyl-1,3-propanediamine/NO (PAPA/NO) and diethylenetriamine/NO (DETA/NO), respectively, via reverse phase evaporation. The anticancer activity of the otherwise equivalent fast and slow NO-releasing systems was evaluated against several distinct pancreatic, colorectal, and breast cancer cell lines. The anticancer assays (via cytotoxicity) over 72 h revealed that the slower NO-releasing liposomes consistently required lower NO payloads (LD50 <3 μg/mL) relative to the fast NO-release system (LD50 >6 μg/mL) to elicit cytotoxicity. The mechanism of intracellular NO build-up in cancer cells was studied using confocal fluorescence microscopy and flow cytometry, the results of which indicated that a more gradual NO accumulation was characteristic of the slow NO-release system. Protein expression via Western blot analysis revealed that slower NO release resulted in more necrotic/apoptotic cells, while faster release reduced the number of mitotic cells to a greater extent. Overall, these studies demonstrate the potential of NO-releasing liposomes for anticancer therapy and highlight the significance of release kinetics (and NO payloads) required to induce cell death.
Collapse
Affiliation(s)
- Dakota J Suchyta
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - Mark H Schoenfisch
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| |
Collapse
|
13
|
Garnacho C, Muro S. ICAM-1 targeting, intracellular trafficking, and functional activity of polymer nanocarriers coated with a fibrinogen-derived peptide for lysosomal enzyme replacement. J Drug Target 2017; 25:786-795. [PMID: 28665212 DOI: 10.1080/1061186x.2017.1349771] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Enzyme replacement is a viable treatment for diseases caused by genetic deficiency of lysosomal enzymes. However, suboptimal access of enzymes to target sites limits this strategy. Polymer nanocarriers (NCs) coated with antibody against intercellular adhesion molecule 1 (ICAM-1), a protein overexpressed on most cells under disease states, enhanced biodistribution and lysosomal delivery of these therapeutics. Whether this can be achieved using more biocompatible ICAM-1-targeting moieties is unknown, since intracellular uptake via this route is sensitive to the receptor epitope being targeted. We examined this using polymer NCs coated with an ICAM-1-targeting peptide derived from the fibrinogen sequence. Scrambled-sequence peptide and anti-ICAM were used as controls. NCs carried acid sphingomyelinase (ASM), used for treatment of type B Niemann-Pick disease, and fluorescence microscopy was employed to examine cellular performance. Peptide-coated/enzyme NCs efficiently targeted ICAM-1 (22-fold over non-specific counterparts; Bmax ∼180 NCs/cell; t1/2 ∼28 min), recognised human and mouse cells (1.2- to 0.7-fold binding vs. antibody/enzyme NCs), were efficiently endocytosed (71% at 1 h chase), and trafficked to lysosomes (30--45% of internalised NCs; 2 h chase). This restored lysosomal levels of sphingomyelin and cholesterol within 5 h chase (∼95% reduction over disease levels), similar to antibody-enzyme NCs. This fibrinogen-derived ICAM-1-targeting peptide holds potential for lysosomal enzyme replacement therapy.
Collapse
Affiliation(s)
- Carmen Garnacho
- a Department of Normal and Pathological Histology and Cytology , University of Seville School of Medicine , Seville , Spain
| | - Silvia Muro
- b Institute for Bioscience & Biotechnology Research, University of Maryland , College Park , MD , USA.,c Fischell Department of Bioengineering , University of Maryland , College Park , MD , USA
| |
Collapse
|
14
|
Ranganathan A, Campo J, Myerson J, Shuvaev V, Zern B, Muzykantov V, Eckmann DM. Fluorescence Microscopy Imaging Calibration for Quantifying Nanocarrier Binding to Cells During Shear Flow Exposure. J Biomed Nanotechnol 2017; 13:737-745. [PMID: 29104516 PMCID: PMC5665578 DOI: 10.1166/jbn.2017.2392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Targeted drug delivery is a fast growing industry in healthcare and technologies are being developed for applications utilizing nanocarriers as vehicles for drug transport. As the size scale of these particles becomes further reduced, advanced fluorescence microscopy and image analysis techniques become increasingly important for facilitating our understanding of nanocarrier binding and avidity, thereby establishing the basis for nanocarrier design optimization. While there is a significant body of published work using nanocarriers in vitro and in vivo, the advent of smaller particles that have typically been studied (~500 nm) limits the ability to attain quantitative measurements of nanocarrier binding dynamics since image acquisition and analysis methods are restricted by microscopy pixel size. This work demonstrates the use of a novel calibration technique based on radioisotope counting and fluorescence imaging for enabling quantitative determination of nanocarrier binding dynamics. The technique is then applied to assess the temporal profile of endothelial cell binding of two antibody targeted nanocarrier types in the presence of fluid shear stress. Results are provided for binding of nanoparticles smaller than a microscopy image pixel.
Collapse
Affiliation(s)
- Abhay Ranganathan
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jessica Campo
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacob Myerson
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vladimir Shuvaev
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Blaine Zern
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vladimir Muzykantov
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David M. Eckmann
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
15
|
Ghaffarian R, Roki N, Abouzeid A, Vreeland W, Muro S. Intra- and trans-cellular delivery of enzymes by direct conjugation with non-multivalent anti-ICAM molecules. J Control Release 2016; 238:221-230. [PMID: 27473764 DOI: 10.1016/j.jconrel.2016.07.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/16/2016] [Accepted: 07/25/2016] [Indexed: 12/27/2022]
Abstract
Intercellular adhesion molecule 1 (ICAM-1) is a cell-surface protein overexpressed in many diseases and explored for endocytosis and transcytosis of drug delivery systems. All previous evidence demonstrating ICAM-1-mediated transport of therapeutics into or across cells was obtained using nanocarriers or conjugates coupled to multiple copies of anti-ICAM antibodies or peptides. Yet, transport of therapeutics linked to non-multivalent anti-ICAM ligands has never been shown, since multivalency was believed to be necessary to induce transport. Our goal was to explore whether non-multivalent binding to ICAM-1 could drive endocytosis and/or transcytosis of model cargo in different cell types. We found that anti-ICAM was specifically internalized by all tested ICAM-1-expressing cells, including epithelial, fibroblast and neuroblastoma cells, primary or established cell lines. Uptake was inhibited at 4°C and in the presence of an inhibitor of the ICAM-1-associated pathway, rather than inhibitors of the clathrin or caveolar routes. We observed minimal transport of anti-ICAM to lysosomes, yet prominent and specific transcytosis across epithelial monolayers. Finally, we coupled a model cargo (the enzyme horseradish peroxidase (HRP)) to anti-ICAM and separated a 1:2 antibody:enzyme conjugate for non-multivalent ICAM-1 targeting. Similar to anti-ICAM, anti-ICAM-HRP was specifically internalized and transported across cells, which rendered intra- and trans-cellular enzyme activity. Therefore, non-multivalent ICAM-1 targeting also provides transport of cargoes into and across cells, representing a new alternative for future therapeutic applications via this route.
Collapse
Affiliation(s)
- Rasa Ghaffarian
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Niksa Roki
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Abraham Abouzeid
- National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Wyatt Vreeland
- National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Silvia Muro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA; Institute of Bioscience & Biotechnology Research, University of Maryland, College Park, MD, USA.
| |
Collapse
|
16
|
Noyhouzer T, L'Homme C, Beaulieu I, Mazurkiewicz S, Kuss S, Kraatz HB, Canesi S, Mauzeroll J. Ferrocene-Modified Phospholipid: An Innovative Precursor for Redox-Triggered Drug Delivery Vesicles Selective to Cancer Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:4169-4178. [PMID: 26987014 DOI: 10.1021/acs.langmuir.6b00511] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Controlled payload release is one of the key elements in the creation of a reliable drug delivery system. We report the discovery of a drug delivery vessel able to transport chemotherapeutic agents to target cancer cells and selectively trigger their release using the electrochemical activity of a ferrocene-modified phospholipid. Supported by in vitro assays, the competitive advantages of this discovery are (i) the simple one step scalability of the synthetic process, (ii) the stable encapsulation of toxic drugs (doxorubicin) during transport, and (iii) the selective redox triggering of the liposomes to harness their cytotoxic payload at the cancer site. Specifically, the redox-modified giant unilamellar vesicle and liposomes were characterized using advanced methods such as scanning electrochemical microscopy (SECM), transmission electron microscopy (TEM), dynamic light scattering (DLS), and fluorescent imaging.
Collapse
Affiliation(s)
- Tomer Noyhouzer
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A OB8
| | - Chloé L'Homme
- Laboratoire de Méthodologie et Synthèse de Produits Naturels, Université du Québec à Montréal , C.P. 8888, Succ. Centre-Ville, Montréal, H3C 3P8, Québec, Canada
| | - Isabelle Beaulieu
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A OB8
| | - Stephanie Mazurkiewicz
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A OB8
| | - Sabine Kuss
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A OB8
| | - Heinz-Bernhard Kraatz
- Department of Physical and Environmental Sciences, University of Toronto Scarborough , Toronto, Ontario M1C 1A4, Canada
| | - Sylvain Canesi
- Laboratoire de Méthodologie et Synthèse de Produits Naturels, Université du Québec à Montréal , C.P. 8888, Succ. Centre-Ville, Montréal, H3C 3P8, Québec, Canada
| | - Janine Mauzeroll
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A OB8
| |
Collapse
|
17
|
Ghaffarian R, Muro S. Distinct subcellular trafficking resulting from monomeric vs multimeric targeting to endothelial ICAM-1: implications for drug delivery. Mol Pharm 2014; 11:4350-62. [PMID: 25301142 PMCID: PMC4255724 DOI: 10.1021/mp500409y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Ligand-targeted,
receptor-mediated endocytosis is commonly exploited for intracellular
drug delivery. However, cells-surface receptors may follow distinct
endocytic fates when bound by monomeric vs multimeric ligands. Our
purpose was to study this paradigm using ICAM-1, an endothelial receptor
involved in inflammation, to better understand its regulation and
potential for drug delivery. Our procedure involved fluorescence microscopy
of human endothelial cells to determine the endocytic behavior of
unbound ICAM-1 vs ICAM-1 bound by model ligands: monomeric (anti-ICAM)
vs multimeric (anti-ICAM biotin–streptavidin conjugates or
anti-ICAM coated onto 100 nm nanocarriers). Our findings suggest that
both monomeric and multimeric ligands undergo a similar endocytic
pathway sensitive to amiloride (∼50% inhibition), but not inhibitors
of clathrin-pits or caveoli. After 30 min, ∼60–70% of
both ligands colocalized with Rab11a-compartments. By 3–5 h,
∼65–80% of multimeric anti-ICAM colocalized with perinuclear
lysosomes with ∼60–80% degradation, while 70% of monomeric
anti-ICAM remained associated with Rab11a at the cell periphery and
recycled to and from the cell-surface with minimal (<10%) lysosomal
colocalization and minimal (≤15%) degradation. In the absence
of ligands, ICAM-1 also underwent amiloride-sensitive endocytosis
with peripheral distribution, suggesting that monomeric (not multimeric)
anti-ICAM follows the route of this receptor. In conclusion, ICAM-1
can mediate different intracellular itineraries, revealing new insight
into this biological pathway and alternative avenues for drug delivery.
Collapse
Affiliation(s)
- Rasa Ghaffarian
- Fischell Department of Bioengineering, University of Maryland , 2330 Jeong H. Kim Engineering Building, College Park, Maryland 20742, United States
| | | |
Collapse
|
18
|
Deddens LH, van Tilborg GAF, van der Toorn A, van der Marel K, Paulis LEM, van Bloois L, Storm G, Strijkers GJ, Mulder WJM, de Vries HE, Dijkhuizen RM. MRI of ICAM-1 upregulation after stroke: the importance of choosing the appropriate target-specific particulate contrast agent. Mol Imaging Biol 2014; 15:411-22. [PMID: 23400400 DOI: 10.1007/s11307-013-0617-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Magnetic resonance imaging (MRI) with targeted contrast agents provides a promising means for diagnosis and treatment monitoring after cerebrovascular injury. Our goal was to demonstrate the feasibility of this approach to detect the neuroinflammatory biomarker intercellular adhesion molecule-1 (ICAM-1) after stroke and to establish a most efficient imaging procedure. PROCEDURES We compared two types of ICAM-1-functionalized contrast agent: T 1-shortening gadolinium chelate-containing liposomes and T2(*)-shortening micron-sized iron oxide particles (MPIO). Binding efficacy and MRI contrast effects were tested in cell cultures and a mouse stroke model. RESULTS Both ICAM-1-targeted agents bound effectively to activated cerebrovascular cells in vitro, generating significant MRI contrast-enhancing effects. Direct in vivo MRI-based detection after stroke was only achieved with ICAM-1-targeted MPIO, although both contrast agents showed similar target-specific vascular accumulation. CONCLUSIONS Our study demonstrates the potential of in vivo MRI of post-stroke ICAM-1 upregulation and signifies target-specific MPIO as most suitable contrast agent for molecular MRI of cerebrovascular inflammation.
Collapse
Affiliation(s)
- Lisette H Deddens
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, Yalelaan 2, 3584 CM, Utrecht, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hua S. Targeting sites of inflammation: intercellular adhesion molecule-1 as a target for novel inflammatory therapies. Front Pharmacol 2013; 4:127. [PMID: 24109453 PMCID: PMC3790104 DOI: 10.3389/fphar.2013.00127] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 09/14/2013] [Indexed: 12/19/2022] Open
Abstract
Targeted drug delivery to sites of inflammation will provide effective, precise, and safe therapeutic interventions for treatment of diverse disease conditions, by limiting toxic side effects and/or increasing drug action. Disease-site targeting is believed to play a major role in the enhanced efficacy observed for a variety of drugs when formulated inside lipid vesicles. This article will focus on the factors and mechanisms involved in drug targeting to sites of inflammation and the importance of cell adhesion molecules, in particular intercellular adhesion molecule-1, in this process.
Collapse
Affiliation(s)
- Susan Hua
- The School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan New South Wales, Australia
| |
Collapse
|
20
|
Ascenso A, Pinho S, Eleutério C, Praça FG, Bentley MVLB, Oliveira H, Santos C, Silva O, Simões S. Lycopene from tomatoes: vesicular nanocarrier formulations for dermal delivery. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:7284-7293. [PMID: 23826819 DOI: 10.1021/jf401368w] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
This experimental work aimed to develop a simple, fast, economic, and environmentally friendly process for the extraction of lycopene from tomato and incorporate this lycopene-rich extract into ultradeformable vesicular nanocarriers suitable for topical application. Lycopene extraction was conducted without a cosolvent for 30 min. The extracts were analyzed and incorporated in transfersomes and ethosomes. These formulations were characterized, and the cellular uptake was observed by confocal microscopy. Dermal delivery of lycopene formulations was tested under in vitro and in vivo conditions. Lycopene extraction proved to be quite safe and selective. The vesicular formulation was taken up by the cells, being more concentrated around the nucleus. Epicutaneous application of lycopene formulations decreased the level of anthralin-induced ear swelling by 97 and 87%, in a manner nonstatistically different from the positive control. These results support the idea that the lycopene-rich extract may be a good alternative to the expensive commercial lycopene for incorporation into advanced topical delivery systems.
Collapse
Affiliation(s)
- Andreia Ascenso
- Nanomedicine and Drug Delivery Systems group of iMedUL, Faculdade de Farmácia da Universidade de Lisboa, Lisboa, Portugal.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Biological Atomic Force Microscopy for Imaging Gold-Labeled Liposomes on Human Coronary Artery Endothelial Cells. JOURNAL OF PHARMACEUTICS 2013; 2013:875906. [PMID: 26555999 PMCID: PMC4590807 DOI: 10.1155/2013/875906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 01/18/2013] [Indexed: 11/21/2022]
Abstract
Although atomic force microscopy (AFM) has been used extensively to characterize cell membrane structure and cellular processes such as endocytosis and exocytosis, the corrugated surface of the cell membrane hinders the visualization of extracellular entities, such as liposomes, that may interact with the cell. To overcome this barrier, we used 90 nm nanogold particles to label FITC liposomes and monitor their endocytosis on human coronary artery endothelial cells (HCAECs) in vitro. We were able to study the internalization process of gold-coupled liposomes on endothelial cells, by using AFM. We found that the gold-liposomes attached to the HCAEC cell membrane during the first 15–30 min of incubation, liposome cell internalization occurred from 30 to 60 min, and most of the gold-labeled liposomes had invaginated after 2 hr of incubation. Liposomal uptake took place most commonly at the periphery of the nuclear zone. Dynasore monohydrate, an inhibitor of endocytosis, obstructed the internalization of the gold-liposomes. This study showed the versatility of the AFM technique, combined with fluorescent microscopy, for investigating liposome uptake by endothelial cells. The 90 nm colloidal gold nanoparticles proved to be a noninvasive contrast agent that efficiently improves AFM imaging during the investigation of biological nanoprocesses.
Collapse
|
22
|
Koning GA, Morselt HW, Kamps JA, Scherphof GL. Uptake and intracellular processing of PEG-liposomes and PEG-immunoliposomes by kupffer cells in vitro 1 *. J Liposome Res 2012; 11:195-209. [PMID: 19530933 DOI: 10.1081/lpr-100108462] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Specific targeting of drugs to for instance tumors or sites of inflammation may be achieved by means of immunoliposomes carrying site-specific antibodies on their surface. The presence of these antibodies may adversely affect the circulation kinetics of such liposomes as a result of interactions with cells of the mononuclear phagocyte system (MPS), mainly represented by macrophages in liver and spleen. The additional insertion of poly(ethylene glycol) chains on the surface of the immunoliposomes may, however, attenuate this effect. We investigated the influence of surface-coupled rat or rabbit antibodies and of PEG on the uptake of liposomes by rat Kupffer cells in culture with (3)H-cholesteryloleyl ether as a metabolically stable marker. Additionally, we assessed the effects of surface-bound IgG and PEG on the intracellular processing of the liposomes by the Kupffer cells, based on a double-label assay using the (3)H-cholesteryl ether as an absolute measure for liposome uptake and the hydrolysis of the degradable marker cholesteryl-(14)C-oleate as relative measure of degradation. Attachment of both rat and rabbit antibodies to PEG-free liposomes caused a several-fold increase in apparent size. The uptake by Kupffer cells, however, was 3-4 fold higher for the rat than for the rabbit IgG liposomes. The presence of PEG drastically reduced the difference between these liposome types. Uptake of liposomes without antibodies amounted to only about 10% (non-PEGylated) or less (PEGylated) of that of the immunoliposomes. In contrast to the marked effects of IgG and PEG on Kupffer cell uptake, the rate of intracellular processing of the liposomes remained virtually unaffected by the presence of these substances on the liposomal surface. These observations are discussed with respect to the design of optimally formulated liposomal drug preparations, combining maximal therapeutic efficacy with minimal toxicity.
Collapse
Affiliation(s)
- G A Koning
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, P. O. Box 80082, 3508, TB, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
23
|
Paulis LEM, Jacobs I, van den Akker NM, Geelen T, Molin DG, Starmans LWE, Nicolay K, Strijkers GJ. Targeting of ICAM-1 on vascular endothelium under static and shear stress conditions using a liposomal Gd-based MRI contrast agent. J Nanobiotechnology 2012; 10:25. [PMID: 22716048 PMCID: PMC3563567 DOI: 10.1186/1477-3155-10-25] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 06/04/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The upregulation of intercellular adhesion molecule-1 (ICAM-1) on the endothelium of blood vessels in response to pro-inflammatory stimuli is of major importance for the regulation of local inflammation in cardiovascular diseases such as atherosclerosis, myocardial infarction and stroke. In vivo molecular imaging of ICAM-1 will improve diagnosis and follow-up of patients by non-invasive monitoring of the progression of inflammation. RESULTS A paramagnetic liposomal contrast agent functionalized with anti-ICAM-1 antibodies for multimodal magnetic resonance imaging (MRI) and fluorescence imaging of endothelial ICAM-1 expression is presented. The ICAM-1-targeted liposomes were extensively characterized in terms of size, morphology, relaxivity and the ability for binding to ICAM-1-expressing endothelial cells in vitro. ICAM-1-targeted liposomes exhibited strong binding to endothelial cells that depended on both the ICAM-1 expression level and the concentration of liposomes. The liposomes had a high longitudinal and transversal relaxivity, which enabled differentiation between basal and upregulated levels of ICAM-1 expression by MRI. The liposome affinity for ICAM-1 was preserved in the competing presence of leukocytes and under physiological flow conditions. CONCLUSION This liposomal contrast agent displays great potential for in vivo MRI of inflammation-related ICAM-1 expression.
Collapse
Affiliation(s)
- Leonie E M Paulis
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Garnacho C, Serrano D, Muro S. A fibrinogen-derived peptide provides intercellular adhesion molecule-1-specific targeting and intraendothelial transport of polymer nanocarriers in human cell cultures and mice. J Pharmacol Exp Ther 2011; 340:638-47. [PMID: 22160267 DOI: 10.1124/jpet.111.185579] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Intercellular adhesion molecule-1 (ICAM-1), a transmembrane glycoprotein expressed on activated endothelium and many other cells, represents a suitable target for delivery of drug nanocarriers (NCs) to disease areas. Numerous works have shown efficient targeting and intracellular transport of ICAM-1-targeted NCs, rendering significant therapeutic potential. This is the case for enzyme delivery for treatment of multitissue lysosomal storage disorders. However, those studies used formulations targeted to ICAM-1 by antibodies (anti-ICAM NCs). This poses an obstacle to preclinical evaluation of long-term treatment of such chronic maladies, caused by immunogenicity of foreign proteins administered to animals, compelling development of alternative strategies. In this work, we used radioisotope tracing, fluorescence and electron microscopy, and in vitro, cell cultures, and mouse models to evaluate polymer nanocarriers targeted to ICAM-1 by a 17-mer linear peptide derived from the ICAM-1-binding sequence of fibrinogen (γ3). Our results show that γ3 NCs target ICAM-1 with efficiency and specificity similar to that of anti-ICAM NCs, determined by using immobilized ICAM-1, native ICAM-1 expressed on endothelial cell cultures, and intravenous administration in mice. Furthermore, γ3 NCs are internalized by cells in culture and in vivo and transported to lysosomes via cell adhesion molecule-mediated endocytosis, without apparent disruption of cell junctions, similar to anti-ICAM counterparts. The degree of conservation of fibrinogen γ3 sequence and its cognate site on ICAM-1 among species (e.g., mouse, chimpanzee, and humans) reflects the interspecies targeting found for γ3 NCs, providing an avenue for exploring the translation of ICAM-1-targeting platforms in the preclinical and, perhaps, future clinical realm.
Collapse
Affiliation(s)
- Carmen Garnacho
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742-4450, USA
| | | | | |
Collapse
|
25
|
Bhowmick T, Berk E, Cui X, Muzykantov VR, Muro S. Effect of flow on endothelial endocytosis of nanocarriers targeted to ICAM-1. J Control Release 2011; 157:485-92. [PMID: 21951807 DOI: 10.1016/j.jconrel.2011.09.067] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 09/10/2011] [Indexed: 01/15/2023]
Abstract
Delivery of drugs into the endothelium by nanocarriers targeted to endothelial determinants may improve treatment of vascular maladies. This is the case for intercellular adhesion molecule 1 (ICAM-1), a glycoprotein overexpressed on endothelial cells (ECs) in many pathologies. ICAM-1-targeted nanocarriers bind to and are internalized by ECs via a non-classical pathway, CAM-mediated endocytosis. In this work we studied the effects of endothelial adaptation to physiological flow on the endocytosis of model polymer nanocarriers targeted to ICAM-1 (anti-ICAM/NCs, ~180 nm diameter). Culturing established endothelial-like cells (EAhy926 cells) and primary human umbilical vein ECs (HUVECs) under 4 dyn/cm(2) laminar shear stress for 24 h resulted in flow adaptation: cell elongation and formation of actin stress fibers aligned to the flow direction. Fluorescence microscopy showed that flow-adapted cells internalized anti-ICAM/NCs under flow, although at slower rate versus non flow-adapted cells under static incubation (~35% reduction). Uptake was inhibited by amiloride, whereas marginally affected by filipin and cadaverine, implicating that CAM-endocytosis accounts for anti-ICAM/NC uptake under flow. Internalization under flow was more modestly affected by inhibiting protein kinase C, which regulates actin remodeling during CAM-endocytosis. Actin recruitment to stress fibers that maintain the cell shape under flow may delay uptake of anti-ICAM/NCs under this condition by interfering with actin reorganization needed for CAM-endocytosis. Electron microscopy revealed somewhat slow, yet effective endocytosis of anti-ICAM/NCs by pulmonary endothelium after i.v. injection in mice, similar to that of flow-adapted cell cultures: ~40% (30 min) and 80% (3 h) internalization. Similar to cell culture data, uptake was slightly faster in capillaries with lower shear stress. Further, LPS treatment accelerated internalization of anti-ICAM/NCs in mice. Therefore, regulation of endocytosis of ICAM-1-targeted nanocarriers by flow and endothelial status may modulate drug delivery into ECs exposed to different physiological (capillaries vs. arterioles/venules) or pathological (ischemia, inflammation) levels and patterns of blood flow.
Collapse
Affiliation(s)
- Tridib Bhowmick
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA
| | | | | | | | | |
Collapse
|
26
|
Hitchcock KE, Caudell DN, Sutton JT, Klegerman ME, Vela D, Pyne-Geithman GJ, Abruzzo T, Cyr PEP, Geng YJ, McPherson DD, Holland CK. Ultrasound-enhanced delivery of targeted echogenic liposomes in a novel ex vivo mouse aorta model. J Control Release 2010; 144:288-95. [PMID: 20202474 PMCID: PMC2878875 DOI: 10.1016/j.jconrel.2010.02.030] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 02/16/2010] [Accepted: 02/25/2010] [Indexed: 11/23/2022]
Abstract
The goal of this study was to determine whether targeted, Rhodamine-labeled echogenic liposomes (Rh-ELIP) containing nanobubbles could be delivered to the arterial wall, and whether 1-MHz continuous wave ultrasound would enhance this delivery profile. Aortae excised from apolipoprotein-E-deficient (n=8) and wild-type (n=8) mice were mounted in a pulsatile flow system through which Rh-ELIP were delivered in a stream of bovine serum albumin. Half the aortae from each group were treated with 1-MHz continuous wave ultrasound at 0.49 MPa peak-to-peak pressure, and half underwent sham exposure. Ultrasound parameters were chosen to promote stable cavitation and avoid inertial cavitation. A broadband hydrophone was used to monitor cavitation activity. After treatment, aortic sections were prepared for histology and analyzed by an individual blinded to treatment conditions. Delivery of Rh-ELIP to the vascular endothelium was observed, and sub-endothelial penetration of Rh-ELIP was present in five of five ultrasound-treated aortae and was absent in those not exposed to ultrasound. However, the degree of penetration in the ultrasound-exposed aortae was variable. There was no evidence of ultrasound-mediated tissue damage in any specimen. Ultrasound-enhanced delivery within the arterial wall was demonstrated in this novel model, which allows quantitative evaluation of therapeutic delivery.
Collapse
Affiliation(s)
- Kathryn E Hitchcock
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hua S, Chang HI, Davies NM, Cabot PJ. Targeting of ICAM-1-directed immunoliposomes specifically to activated endothelial cells with low cellular uptake: use of an optimized procedure for the coupling of low concentrations of antibody to liposomes. J Liposome Res 2010; 21:95-105. [PMID: 20429814 DOI: 10.3109/08982101003754401] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Targeted delivery of therapeutics to the endothelium is an important goal in the treatment of inflammatory diseases. The aim of this work was to exploit the overexpression of intercellular adhesion molecule-1 (ICAM-1) on activated endothelial cells for the targeting of anti-ICAM-1-coupled immunoliposomes with the intent for further use as drug carriers. Immunoliposomes were prepared from using an optimized method for the coupling of low concentrations of antibody to liposomes, thereby preventing the loss of antibody through the derivatization, extraction, and activation process. This is especially suitable for limiting ligand conjugates that are isolated or synthesized in small quantities, such as monoclonal antibodies (mAbs). To investigate the functionality of the resulting immunoliposomes, the specificity of binding and cellular internalization studies of liposomes, either nonconjugated or conjugated with mAbs to ICAM-1 or to irrelevant IgG to high endothelial venule (HEV) cells, were analyzed by fluorescence microplate spectroscopy at 4 and 37°C. Immunoliposomes specifically directed against ICAM-1 were shown to bind selectively and specifically to tumor necrosis factor alpha-activated endothelial cells in vitro, with minimal cellular internalization. This study provides a novel delivery system that has the potential for targeting therapeutics to inflammatory tissue.
Collapse
Affiliation(s)
- Susan Hua
- The School of Pharmacy, The University of Queensland, Brisbane, Australia
| | | | | | | |
Collapse
|
28
|
Inhibition of breast cancer metastasis by dual liposomes to disturb complex formation. Int J Pharm 2008; 370:121-8. [PMID: 19100823 DOI: 10.1016/j.ijpharm.2008.11.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 11/21/2008] [Accepted: 11/24/2008] [Indexed: 11/22/2022]
Abstract
The interaction between tumour cells and blood components, mainly platelets, plays an important role in metastasis. In this study, the anti-metastatic effect of vesicles containing the cytotoxic drug perifosine (OPP) and the haemostatic inhibitor dipyridamole (DIP) was tested. These dual liposomes (DIP/OPP-L) encapsulating up to 400microg DIP/ml and 6mM OPP were prepared by extrusion technique. In vitro, DIP/OPP-L significantly inhibited the aggregation of platelets and reduced their adhesion to immobilized MT3 cells by up to 60% and 24.7%, respectively. Complex formation between platelets and tumour cells in vitro was completely prevented by DIP/OPP-vesicles. These combinatory liposomes also inhibited the metastatic capacity of circulating tumour cells by reducing the complex formation with platelets. Formation of lung and extrapulmonary metastases after intravenous administration of MT3 breast cancer cells was significantly reduced when mice were treated with a single intravenous dose of DIP/OPP-L containing 100nmol lipid 6h before tumour cell inoculation.
Collapse
|
29
|
VanAuker MD, Hood E. Delivery strategies to target therapies to inflammatory tissue. Expert Opin Drug Deliv 2008; 5:767-74. [PMID: 18590461 DOI: 10.1517/17425247.5.7.767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Inflammation plays a key role in many chronic disease processes as well as an acute role in injury and wound healing. Various cell types are recruited from the bloodstream to the inflamed site through adhesion molecules, cytokines, chemokines and others. OBJECTIVES This review examines many drug-targeting strategies that make use of these molecules or signaling pathways, and seeks to describe certain commonalities irrespective of the disease process or agent to be delivered. METHODS A survey of the literature, primarily within the last year, was performed. Search words included 'drug targeting' and 'inflammation' and of those, the scope was refined to include those studies that specifically sought to modify or ameliorate an aspect of the inflammatory process in the treatment of a disease. RESULTS/CONCLUSION Inflammation plays a key role in many diseases, and many similar targets (such as adhesion molecules) are the focus of the treatment of those diseases.
Collapse
Affiliation(s)
- Michael D VanAuker
- Department of Chemical and Biomedical Engineering, University of South Florida, 4202 E. Fowler Avenue, Tampa, FL 33620, USA.
| | | |
Collapse
|
30
|
Visser CC, Voorwinden LH, Harders LR, Eloualid M, van Bloois L, Crommelin DJA, Danhof M, de Boer AG. Coupling of Metal Containing Homing Devices to Liposomes via a Maleimide Linker: Use of TCEP to Stabilize Thiol-groups without Scavenging Metals. J Drug Target 2008; 12:569-73. [PMID: 15621682 DOI: 10.1080/10611860400010689] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Liposomes for drug delivery are often prepared with maleimide groups on the distal end of PEG to enable coupling of homing devices, such as antibodies, or other proteins. EDTA is used to stabilize the thiol group in the homing device for attachment to the maleimide. However, when using a homing device that contains a metal, EDTA inactivates this by scavenging of the metal. Holo-transferrin (Tf) containing two iron atoms (Fe(3+)), has a much higher affinity for the Tf receptor than apo-Tf (which does not contain any Fe(3+)). To couple Tf to a liposome, the introduction of a thiol group is necessary. During this process, by using N-succinimidyl S-acetylthioacetate (SATA), followed by 2-3 h coupling to the liposomes, Fe(3+) is scavenged by EDTA. This causes a decreased affinity of Tf for its receptor, resulting in a decreased targeting efficiency of the liposomes. Tris(2-carboxyethyl)phosphine (TCEP) hydrochloride is a sulfhydryl reductant that is often used in protein biochemistry. We found that TCEP (0.01 mM) does not scavenge Fe(3+) from Tf and is able to protect thiol groups for the coupling to maleimide. Furthermore, TCEP does not interfere with the maleimide coupling itself. In this communication, we describe the preparation of liposomes, focussing on the coupling of Tf to the maleimide linker at the distal end of PEG, without loosing Fe(3+) from Tf. This method can be applied to other metal-containing homing devices as well.
Collapse
Affiliation(s)
- Corine C Visser
- Leiden/Amsterdam Center for Drug Research (LACDR), Leiden University, Division of Pharmacology, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Simone E, Ding BS, Muzykantov V. Targeted delivery of therapeutics to endothelium. Cell Tissue Res 2008; 335:283-300. [PMID: 18815813 DOI: 10.1007/s00441-008-0676-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Accepted: 08/18/2008] [Indexed: 12/27/2022]
Abstract
The endothelium is a target for therapeutic and diagnostic interventions in a plethora of human disease conditions including ischemia, inflammation, edema, oxidative stress, thrombosis and hemorrhage, and metabolic and oncological diseases. Unfortunately, drugs have no affinity to the endothelium, thereby limiting the localization, timing, specificity, safety, and effectiveness of therapeutic interventions. Molecular determinants on the surface of resting and pathologically altered endothelial cells, including cell adhesion molecules, peptidases, and receptors involved in endocytosis, can be used for drug delivery to the endothelial surface and into intracellular compartments. Drug delivery platforms such as protein conjugates, recombinant fusion constructs, targeted liposomes, and stealth polymer carriers have been designed to target drugs and imaging agents to these determinants. We review endothelial target determinants and drug delivery systems, describe parameters that control the binding of drug carriers to the endothelium, and provide examples of the endothelial targeting of therapeutic enzymes designed for the treatment of acute vascular disorders including ischemia, oxidative stress, inflammation, and thrombosis.
Collapse
Affiliation(s)
- Eric Simone
- Department of Bioengineering, Program in Targeted Therapeutics of Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Engineering and Applied Sciences, Philadelphia, PA, USA
| | | | | |
Collapse
|
32
|
Lorz BG, Smith AS, Gege C, Sackmann E. Adhesion of giant vesicles mediated by weak binding of sialyl-LewisX to E-selectin in the presence of repelling poly(ethylene glycol) molecules. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2007; 23:12293-12300. [PMID: 17918980 DOI: 10.1021/la701824q] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Prior to establishing tight contact with the endothelium, cells such as leukocytes or cancer cells use the recognition between sialyl-LewisX ligands and E-selectin receptors to establish weak, reversible adhesion and to roll along the vessel wall. We study the physical aspects of this process by constructing a mimetic system that consists of a giant fluid vesicle with incorporated lipid-anchored sialyl-LewisX molecules that bind to E-selectin that is immobilized on the flat substrate. The vesicles also carry a certain fraction of repelling PEG2000 molecules. We analyze the equilibrium state of adhesion in detail by means of reflection interference contrast microscopy and find that the adhesion process relies purely on the formation of one or more adhesion domains within the vesicle-substrate contact zone. We find that the content of ligands in the vesicle must be above 5 mol % to establish specific contacts. All concentrations of sialyl-LewisX above 8 mol % provide a very similar final state of adhesion. However, the size and shape of the adhesion domains strongly depend on both the concentrations of E-selectin (0-3500 molecules/microm2) and PEG2000 (0-5 mol %). At 3500 E-selectin molecules/microm2 and small concentrations of PEG2000, the vesicle-substrate contact is maximized and fully occupied by a single adhesion domain. At concentrations of 5 mol %, PEG2000 completely impedes the specific binding to any substrate. Lastly, an increase in the adhesion strength is observed in systems with identical compositions if the reduced volume of the vesicles is larger.
Collapse
Affiliation(s)
- Barbara G Lorz
- E22 Institut für Biophysik, Technische Universität München, D-85748 Garching, Germany
| | | | | | | |
Collapse
|
33
|
Hood E, Gonzalez M, Plaas A, Strom J, VanAuker M. Immuno-targeting of nonionic surfactant vesicles to inflammation. Int J Pharm 2007; 339:222-30. [PMID: 17448616 DOI: 10.1016/j.ijpharm.2006.12.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 12/21/2006] [Accepted: 12/22/2006] [Indexed: 12/24/2022]
Abstract
Niosomes composed of sorbitan monostearate (Span 60), polyoxyethylene sorbitan monostearate (Tween 61), cholesterol, and dicetyl phosphate were conjugated with a purified monoclonal antibody to CD44 (IM7) through a cyanuric chloride (CC) linkage on the polyoxyethylene group of the Tween 61 molecule. Inclusion of small amounts of Tween 61 within the surfactant component of niosomes formed using thin film hydration techniques and sonication did not hamper vesicle stability as compared to Span 60 niosomes. Conjugation was verified by UV absorbance of fluorescently tagged IM7 in non-fluorescing niosomes and fluorescent micrographs. The immuno-niosomes were incubated with synovial lining cells expressing CD44. Attachment of niosomes was evident and showed selectivity and specificity compared to controls. These findings suggest that the resulting immuno-niosomes may provide an effective method for targeted drug delivery.
Collapse
Affiliation(s)
- Elizabeth Hood
- Biomedical Engineering Program, Department of Chemical Engineering, University of South Florida, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
34
|
Abstract
This review focuses on the therapeutic utility of liposomes in the treatment of inflammatory disorders, and aims to offer the reader an overview of the in vivo results obtained with liposomally encapsulated anti-inflammatory and immune suppressive drugs. The past 30 years has clearly indicated the added value of liposomes in the search for solutions for the delivery problems encountered. However, only a few liposomal anti-inflammatory therapeutics have entered the clinic. Reasons for the hurdles existing in the translation of promising preclinical findings to clinical studies are discussed.
Collapse
Affiliation(s)
- Josbert M Metselaar
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, PO Box 80082, 3508 TB Utrecht, The Netherlands
| | | |
Collapse
|
35
|
Voinea M, Manduteanu I, Dragomir E, Capraru M, Simionescu M. Immunoliposomes directed toward VCAM-1 interact specifically with activated endothelial cells--a potential tool for specific drug delivery. Pharm Res 2005; 22:1906-17. [PMID: 16088429 DOI: 10.1007/s11095-005-7247-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2005] [Accepted: 07/05/2005] [Indexed: 02/02/2023]
Abstract
PURPOSE Immunoliposomes can be potentially used as carriers for drug delivery to specific cells. The aim of this paper was to exploit the overexpression of vascular cell adhesion molecule-1 (VCAM-1) on activated human endothelial cells (HEC) for targeting of anti-VCAM-1 coupled liposomes with the intent for further use as drug carriers. METHODS TNF-alpha-activated HEC were exposed to liposomes, either plain or coupled with antibodies to VCAM-1 (L-VCAM-1) or to irrelevant IgG (L-IgG); nonactivated HEC subjected to the same conditions were used as control. For binding studies, the cells were incubated with fluorescently labeled liposomes at 4 degrees C, and after 2 h, fluorescence intensity was assessed by flow cytometry; specificity of binding was determined by performing the experiments in the presence of excess anti-VCAM-1. Cellular internalization of liposomes was studied employing radioactively or fluorescently labelled liposomes; to detect the mechanisms of uptake, experiments were performed in the presence of agents that interfere in the endocytotic pathway. Transmigration of liposomes was monitored in a two-chamber culture model. The effect of L-VCAM-1 binding to HEC on intracellular calcium ([Ca(2+)](i)) and distribution of actin was determined by fluorimetry and fluorescence microscopy. RESULTS (1) L-VCAM-1 binds selectively and specifically to TNF-alpha activated HEC. (2) Approximately 50% of L-VCAM-1 is taken up by receptor-mediated endocytosis via clathrin-coated vesicles. (3) Binding of L-VCAM-1 to HEC surface induces a rise in [Ca(2+)](i) and reorganization of actin filaments. (4) A small percentage of liposomes migrates across HEC. CONCLUSION The data indicate that VCAM-1 may be an appropriate target for specific drug delivery to activated HEC using immunoliposomes.
Collapse
Affiliation(s)
- Manuela Voinea
- Institute of Cellular Biology and Pathology, Bucharest, Romania.
| | | | | | | | | |
Collapse
|
36
|
Cryan SA. Carrier-based strategies for targeting protein and peptide drugs to the lungs. AAPS JOURNAL 2005; 7:E20-41. [PMID: 16146340 PMCID: PMC2751494 DOI: 10.1208/aapsj070104] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
With greater interest in delivery of protein and peptide-based drugs to the lungs for topical and systemic activity, a range of new devices and formulations are being investigated. While a great deal of recent research has focused on the development of novel devices, attention must now be paid to the formulation of these macromolecular drugs. The emphasis in this review will be on targeting of protein/peptide drugs by inhalation using carriers and ligands.
Collapse
Affiliation(s)
- Sally-Ann Cryan
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| |
Collapse
|
37
|
Muro S, Gajewski C, Koval M, Muzykantov VR. ICAM-1 recycling in endothelial cells: a novel pathway for sustained intracellular delivery and prolonged effects of drugs. Blood 2004; 105:650-8. [PMID: 15367437 DOI: 10.1182/blood-2004-05-1714] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is a target for drug delivery to endothelial cells (ECs), which internalize multivalent anti-ICAM nanocarriers (anti-ICAM/NCs) within 15 to 30 minutes. The concomitant ICAM-1 disappearance from the EC surface transiently inhibited subsequent binding and uptake of anti-ICAM/NCs. Within 1 hour, internalized ICAM-1 diverged from anti-ICAM/NCs into prelysosomal vesicles, resurfaced, and enabled uptake of a subsequent anti-ICAM/NC dose. Thus, internalized ICAM-1 was able to recycle back to the plasma membrane. In vivo pulmonary targeting of a second anti-ICAM/NC dose injected 15 minutes after the first dose was decreased by 50% but recovered between 30 minutes and 2.5 hours, comparable to cultured ECs. Anti-ICAM/NCs affected neither EC viability nor fluid-phase endocytosis and traffic to lysosomes. However, lysosomal trafficking of the second dose of anti-ICAM/NCs was decelerated at least 2-fold versus the first dose; hence the major fraction of anti-ICAM/NCs resided in prelysosomal vesicles for at least 5 hours without degradation. Two successive doses of anti-ICAM/NC/catalase protected ECs against H2O2 for at least 8 hours versus 2 hours afforded by a single dose, suggesting that recurrent targeting to ICAM-1 affords longer effects. ICAM-1 recycling and inhibited lysosomal traffic/degradation of subsequent doses may help to prolong activity of therapeutic agents delivered into ECs by anti-ICAM/NCs.
Collapse
Affiliation(s)
- Silvia Muro
- Institute for Environmental Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6068, USA.
| | | | | | | |
Collapse
|
38
|
Carrion C, de Madariaga MA, Domingo JC. In vitro cytotoxic study of immunoliposomal doxorubicin targeted to human CD34+ leukemic cells. Life Sci 2004; 75:313-28. [PMID: 15135652 DOI: 10.1016/j.lfs.2003.12.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2003] [Accepted: 12/11/2003] [Indexed: 10/26/2022]
Abstract
The expression of CD34 antigen in acute myelogenous leukemias is considered an unfavourable prognosis marker for response to anticancer drugs and duration of remission. This study investigated the applicability of long-circulating immunoliposomes loaded with doxorubicin targeted to CD34 antigen present on MDR(+) human myelogenous leukemia KG-1a cell line. Immunoliposomal doxorubicin showed a higher cytotoxicity against KG-1a cells than non-targeted liposomal doxorubicin, but it did not improve over that of free drug. Although no reversal of doxorubicin resistance was found to occur through its liposomal encapsulation, a therapeutic benefit can be obtained by the selective cytotoxicity observed. Endocytosis studies demonstrated that, after binding to CD34 antigen, the immunoliposomes are not internalized by the KG-1a cells and so the cytotoxic effect might be due to drug released into the space near the cell membrane. Thus, immunotargeting of liposomal doxorubicin to CD34(+) leukemic cells may only provide an ex vivo strategy for site-selective CD34(+) leukemia cell killing.
Collapse
MESH Headings
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/pharmacokinetics
- Antibiotics, Antineoplastic/pharmacology
- Antibodies, Monoclonal
- Antigens, CD34/immunology
- Capsules
- Cell Division
- Cell Line, Tumor
- Cell Survival/drug effects
- Chemistry, Pharmaceutical
- Doxorubicin/administration & dosage
- Doxorubicin/pharmacokinetics
- Doxorubicin/pharmacology
- Drug Carriers
- Drug Delivery Systems
- Endocytosis/drug effects
- Flow Cytometry
- Humans
- Immunochemistry
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Liposomes
- Microscopy, Confocal
- Particle Size
Collapse
Affiliation(s)
- C Carrion
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, University of Barcelona. Martí i Franqués 1, 08028 Barcelona, Spain
| | | | | |
Collapse
|
39
|
Zeisig R, Stahn R, Wenzel K, Behrens D, Fichtner I. Effect of sialyl Lewis X-glycoliposomes on the inhibition of E-selectin-mediated tumour cell adhesion in vitro. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2004; 1660:31-40. [PMID: 14757218 DOI: 10.1016/j.bbamem.2003.10.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The aim of this study was to evaluate the potential of different types of sialyl Lewis X-conjugated liposomes as competitive inhibitors for tumour cell adhesion to endothelial E-selectin. Sterically stabilised liposomes with the sLeX ligand at the terminal end of the polyethyleneglycol (PEG) chain, as well as vesicles that had the ligand embedded within the PEG-layer, were compared to ligand-bearing liposomes without sterical stabilisation. First, 14 different tumour cell lines were characterised for their expression of sialyl Lewis X and/or A. Tumour cell adhesion was characterised in three static assays in vitro using: (i) immobilised E-selectin, (ii) CHO cells, transfected to express E-selectin and (iii) human umbilical vein endothelial cells (HUVEC). Sterically stabilised liposomes with the ligand at the terminal end of the polyethylene chain were the most effective inhibitors in all three assays and inhibited the adhesion of HT29 colon- and Lewis lung (LL) carcinoma cells by about 60-80%. The binding was not affected by a PEG-coating of the liposomes. Sterical stabilisation, on the other hand, completely prevented macrophage uptake (J774 cell line) independently of the presence of the ligand, while plain liposomes were taken up in an amount of 5.4 nmol liposomal lipids/10(6) macrophages.
Collapse
Affiliation(s)
- Reinhard Zeisig
- Max-Delbrück-Center for Molecular Medicine, 13092 Berlin-Buch, PF 74 02 38 AG Experimental Pharmacology, 13125 Berlin, Germany.
| | | | | | | | | |
Collapse
|
40
|
Chiu GNC, Bally MB, Mayer LD. Targeting of antibody conjugated, phosphatidylserine-containing liposomes to vascular cell adhesion molecule 1 for controlled thrombogenesis. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1613:115-21. [PMID: 12832092 DOI: 10.1016/s0005-2736(03)00142-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Phosphatidylserine (PS) membrane exposure plays an important role in blood coagulation, and the development of a liposome formulation containing PS may be of potential therapeutic utility if they can be designed to achieve tumor selective thrombosis. The objective of this study was to develop proof-of-principle data for a thrombogenic PS liposome targeted to vascular cell adhesion molecule 1 (VCAM-1) via the attachment of an anti-VCAM-1 monoclonal antibody (Ab). We have evaluated binding of the anti-VCAM-1 Ab-conjugated PS liposomes to VCAM-1 using two in vitro models, as well as assessing the ability of these liposomes to catalyze blood coagulation reactions. Binding of the Ab-conjugated PS liposomes containing 2 or 14 mol% 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[poly(ethylene glycol) 2000] (DSPE-PEG(2000)) to interleukin 1alpha stimulated human umbilical vein endothelial cells was 8- and 16-fold higher than those without conjugated Ab, respectively, based on the percentage relative increase in cell associated lipid for these liposomes. Binding to VCAM-1-coated ELISA plates produced similar results. The VCAM-1-bound Ab-conjugated PS liposomes were capable of catalyzing blood coagulation reactions upon the exposure of the thrombogenic PS membrane surface. This control of PS surface exposure was achieved using exchangeable PEG-derivatized phosphatidylethanolamines (PE-PEG), with 97% of clotting activity recovered after PE-PEG exchanged out. Our results demonstrate the potential for considering further development of procoagulant liposomes that selectively target thrombogenesis in tumor vasculature.
Collapse
Affiliation(s)
- Gigi N C Chiu
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver, BC, Canada
| | | | | |
Collapse
|
41
|
Murciano JC, Muro S, Koniaris L, Christofidou-Solomidou M, Harshaw DW, Albelda SM, Granger DN, Cines DB, Muzykantov VR. ICAM-directed vascular immunotargeting of antithrombotic agents to the endothelial luminal surface. Blood 2003; 101:3977-84. [PMID: 12531816 DOI: 10.1182/blood-2002-09-2853] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Drug targeting to a highly expressed, noninternalizable determinant up-regulated on the perturbed endothelium may help to manage inflammation and thrombosis. We tested whether inter-cellular adhesion molecule-1 (ICAM-1) targeting is suitable to deliver antithrombotic drugs to the pulmonary vascular lumen. ICAM-1 antibodies bind to the surface of endothelial cells in culture, in perfused lungs, and in vivo. Proinflammatory cytokines enhance anti-ICAM binding to the endothelium without inducing internalization. (125)I-labeled anti-ICAM and a reporter enzyme (beta-Gal) conjugated to anti-ICAM bind to endothelium and accumulate in the lungs after intravenous administration in rats and mice. Anti-ICAM is seen to localize predominantly on the luminal surface of the pulmonary endothelium by electron microscopy. We studied the pharmacological effect of ICAM-directed targeting of tissue-type plasminogen activator (tPA). Anti-ICAM/tPA, but not control IgG/tPA, conjugate accumulates in the rat lungs, where it exerts plasminogen activator activity and dissolves fibrin microemboli. Therefore, ICAM may serve as a target for drug delivery to endothelium, for example, for pulmonary thromboprophylaxis. Enhanced drug delivery to sites of inflammation and the potential anti-inflammatory effect of blocking ICAM-1 may enhance the benefit of this targeting strategy.
Collapse
Affiliation(s)
- Juan-Carlos Murciano
- Institute of Environmental Medicine, Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Koning GA, Schiffelers RM, Storm G. Endothelial cells at inflammatory sites as target for therapeutic intervention. ENDOTHELIUM : JOURNAL OF ENDOTHELIAL CELL RESEARCH 2003; 9:161-71. [PMID: 12380641 DOI: 10.1080/10623320213631] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In the course of an inflammation, vascular endothelial cells (VECs) are strongly involved in processes like leukocyte recruitment, cytokine production, and angiogenesis. Specific interference in these processes may yield great therapeutic benefit in the treatment of (chronic) inflammatory disorders. Drug targeting to VECs at inflamed sites may allow such intervention. VECs at inflamed sites represent a very well-accessible target cell population for circulating drug-targeting systems, which may also be selectively distinguished from normal VECs by the expression of several cell surface receptors involved in the inflammation. One group of specifically expressed molecules are the adhesion molecules (AMs), which have a major function in adhesion of cells to each other, to the extracellular matrix, or in the adhesion and subsequent recruitment of circulating immune cells. This review describes AMs with regard to their function in the inflammatory disease and their usefulness in functioning as a specific target receptor for drug-targeting approaches in general and with an emphasis on liposome-based drug delivery.
Collapse
Affiliation(s)
- Gerben A Koning
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Utrecht University, The Netherlands.
| | | | | |
Collapse
|
43
|
Kessner S, Krause A, Rothe U, Bendas G. Investigation of the cellular uptake of E-Selectin-targeted immunoliposomes by activated human endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1514:177-90. [PMID: 11557019 DOI: 10.1016/s0005-2736(01)00368-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the present study the cellular uptake of targeted immunoliposomes by interleukin-1 activated human endothelial cells has been analysed by several spectroscopical and microscopical fluorescence techniques. Previous in vitro experiments demonstrated that the targeting of immunoliposomes to vascular selectins is a potential way for a selective drug delivery at inflammatory sites. In attempts to further adapt the targeting experiments to physiological conditions, we demonstrate that E-Selectin-directed immunoliposomes are able to bind their target cells under the simulated shear force conditions of capillary blood flow cumulatively for up to 18 h. In order to consequently follow the fate of liposomes after target binding, we analysed the route and degree of liposome internalization of the cells concentrating on cell activation state or various liposomal parameters, e.g., sterical stabilization, type of antibody or antibody coupling strategy. The use of NBD-labelled liposomes and subsequent fluorescence quenching outside the cells with dithionite show that circa 25% of the targeted immunoliposomes were internalized. According to inhibition experiments with agents that interfered with the endocytotic pathway, we found out that the major mechanism of liposome entry is endocytic. The entry involves, at least in part, receptor-mediated endocytosis via E-Selectin, a liposome accumulation in the endosomes and their acidification was proved by pyranine spectroscopic results. Furthermore, microscopical investigations demonstrate that also a fusion of liposomes with the cell membrane occurs followed by a release of entrapped calcein into the cytoplasm. These observations gain insight into the behaviour of E-Selectin-targeted immunoliposomes and indicate that these immunoliposomes have great potential to be used as drug carriers for intracellular drug delivery at inflammatory sites.
Collapse
Affiliation(s)
- S Kessner
- Departement of Pharmacy, Martin-Luther-University Halle, Germany
| | | | | | | |
Collapse
|
44
|
Abstract
The incidence of chronic obstructive pulmonary disease (COPD) is increasing throughout the world. Much less is known about the pathogenesis of COPD than that of asthma and there is little response to current therapy. Most patients with COPD have acquired their lung disease through smoking cigarettes, and the major step in management is to minimise further damage by stopping this habit. A number of therapies are being developed for the treatment of COPD; including new bronchodilators such as tiotropium bromide, agents to block inflammation induced by neutrophils and macrophages, as well as strategies to combat proteases and oxidants. The long-term goal is to provide therapy that retards the accelerated loss of lung function occurring in COPD. Development of novel therapies for COPD requires reliable Phase II decision making before entering large scale Phase III studies. The patient with COPD is often overlooked compared to their asthmatic counterpart, who benefit from an urgent need to identify novel targets and better therapy.
Collapse
Affiliation(s)
- M J Leckie
- National Heart and Lung Institute, Royal Brompton Clinical Studies Unit, Imperial College, London, UK
| | | | | | | |
Collapse
|
45
|
Mastrobattista E, Koning GA, Storm G. Immunoliposomes for the targeted delivery of antitumor drugs. Adv Drug Deliv Rev 1999; 40:103-127. [PMID: 10837783 DOI: 10.1016/s0169-409x(99)00043-5] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This review presents an overview of the field of immunoliposome-mediated targeting of anticancer agents. First, problems that are encountered when immunoliposomes are used for systemic anticancer drug delivery and potential solutions are discussed. Second, an update is given of the in vivo results obtained with immunoliposomes in tumor models. Finally, new developments on the utilization of immunoliposomes for the treatment of cancer are highlighted.
Collapse
Affiliation(s)
- E Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Pharmacy, Utrecht University, Sorbonnelaan 16, 3508 TB, Utrecht, The Netherlands
| | | | | |
Collapse
|