1
|
Torres-Herrero B, Armenia I, Ortiz C, de la Fuente JM, Betancor L, Grazú V. Opportunities for nanomaterials in enzyme therapy. J Control Release 2024; 372:619-647. [PMID: 38909702 DOI: 10.1016/j.jconrel.2024.06.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
In recent years, enzyme therapy strategies have rapidly evolved to catalyze essential biochemical reactions with therapeutic potential. These approaches hold particular promise in addressing rare genetic disorders, cancer treatment, neurodegenerative conditions, wound healing, inflammation management, and infectious disease control, among others. There are several primary reasons for the utilization of enzymes as therapeutics: their substrate specificity, their biological compatibility, and their ability to generate a high number of product molecules per enzyme unit. These features have encouraged their application in enzyme replacement therapy where the enzyme serves as the therapeutic agent to rectify abnormal metabolic and physiological processes, enzyme prodrug therapy where the enzyme initiates a clinical effect by activating prodrugs, and enzyme dynamic or starving therapy where the enzyme acts upon host substrate molecules. Currently, there are >20 commercialized products based on therapeutic enzymes, but approval rates are considerably lower than other biologicals. This has stimulated nanobiotechnology in the last years to develop nanoparticle-based solutions that integrate therapeutic enzymes. This approach aims to enhance stability, prevent rapid clearance, reduce immunogenicity, and even enable spatio-temporal activation of the therapeutic catalyst. This comprehensive review delves into emerging trends in the application of therapeutic enzymes, with a particular emphasis on the synergistic opportunities presented by incorporating enzymes into nanomaterials. Such integration holds the promise of enhancing existing therapies or even paving the way for innovative nanotherapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Torres-Herrero
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Cecilia Ortiz
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Jesús Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Lorena Betancor
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
2
|
Qin H, Teng Y, Dai R, Wang A, Liu J. Glycan-based scaffolds and nanoparticles as drug delivery system in cancer therapy. Front Immunol 2024; 15:1395187. [PMID: 38799466 PMCID: PMC11116596 DOI: 10.3389/fimmu.2024.1395187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Glycan-based scaffolds are unique in their high specificity, versatility, low immunogenicity, and ability to mimic natural carbohydrates, making them attractive candidates for use in cancer treatment. These scaffolds are made up of glycans, which are biopolymers with well biocompatibility in the human body that can be used for drug delivery. The versatility of glycan-based scaffolds allows for the modulation of drug activity and targeted delivery to specific cells or tissues, which increases the potency of drugs and reduces side effects. Despite their promise, there are still technical challenges in the design and production of glycan-based scaffolds, as well as limitations in their therapeutic efficacy and specificity.
Collapse
Affiliation(s)
- Henan Qin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yibin Teng
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Rui Dai
- Department of Pharmacy, Peking Union Medical University Hospital, Beijing, China
| | - Aman Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
3
|
Melrose J. Hyaluronan hydrates and compartmentalises the CNS/PNS extracellular matrix and provides niche environments conducive to the optimisation of neuronal activity. J Neurochem 2023; 166:637-653. [PMID: 37492973 DOI: 10.1111/jnc.15915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
The central nervous system/peripheral nervous system (CNS/PNS) extracellular matrix is a dynamic and highly interactive space-filling, cell-supportive, matrix-stabilising, hydrating entity that creates and maintains tissue compartments to facilitate regional ionic micro-environments and micro-gradients that promote optimal neural cellular activity. The CNS/PNS does not contain large supportive collagenous and elastic fibrillar networks but is dominated by a high glycosaminoglycan content, predominantly hyaluronan (HA) and collagen is restricted to the brain microvasculature, blood-brain barrier, neuromuscular junction and meninges dura, arachnoid and pia mater. Chondroitin sulphate-rich proteoglycans (lecticans) interactive with HA have stabilising roles in perineuronal nets and contribute to neural plasticity, memory and cognitive processes. Hyaluronan also interacts with sialoproteoglycan associated with cones and rods (SPACRCAN) to stabilise the interphotoreceptor matrix and has protective properties that ensure photoreceptor viability and function is maintained. HA also regulates myelination/re-myelination in neural networks. HA fragmentation has been observed in white matter injury, multiple sclerosis, and traumatic brain injury. HA fragments (2 × 105 Da) regulate oligodendrocyte precursor cell maturation, myelination/remyelination, and interact with TLR4 to initiate signalling cascades that mediate myelin basic protein transcription. HA and its fragments have regulatory roles over myelination which ensure high axonal neurotransduction rates are maintained in neural networks. Glioma is a particularly invasive brain tumour with extremely high mortality rates. HA, CD44 and RHAMM (receptor for HA-mediated motility) HA receptors are highly expressed in this tumour. Conventional anti-glioma drug treatments have been largely ineffective and surgical removal is normally not an option. CD44 and RHAMM glioma HA receptors can potentially be used to target gliomas with PEP-1, a cell-penetrating HA-binding peptide. PEP-1 can be conjugated to a therapeutic drug; such drug conjugates have successfully treated dense non-operative tumours in other tissues, therefore similar applications warrant exploration as potential anti-glioma treatments.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
- Sydney Medical School, Northern, The University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| |
Collapse
|
4
|
Kumari S, Choudhary PK, Shukla R, Sahebkar A, Kesharwani P. Recent advances in nanotechnology based combination drug therapy for skin cancer. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:1435-1468. [PMID: 35294334 DOI: 10.1080/09205063.2022.2054399] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Skin-cancer (SC) is more common than all other cancers affecting large percentage of the population in the world and is increasing in terms of morbidity and mortality. In the United States, 3million people are affected by SC annually whereas millions of people are affected globally. Melanoma is fifth most common cancer in the United States. SC is commonly occurred in white people as per WHO. SC is divided into two groups, i.e. melanoma and non-melanoma. In the previous two decades, management of cancer remains to be a tough and a challenging task for many scholars. Presently, the treatment protocols are mostly based on surgery and chemo-radiation therapy, which sooner or later harm the unaffected cells too. To reduce these limitations, nano scaled materials and its extensive range may be recognized as the probable carriers for the selective drug delivery in response to cancerous cells. Recently, the nanocarriers based drugs and their combinations were found to be a new and interesting approach of study for the management of skin carcinoma to enhance the effectiveness, to lessen the dose-dependent side effects and to avoid the drug resistance. This review may emphasize on the wide-range of information on nanotechnology-based drugs and their combination with physical techniques.
Collapse
Affiliation(s)
- Shweta Kumari
- Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, India
| | | | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P., India
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
5
|
Diniz F, Coelho P, Duarte HO, Sarmento B, Reis CA, Gomes J. Glycans as Targets for Drug Delivery in Cancer. Cancers (Basel) 2022; 14:cancers14040911. [PMID: 35205658 PMCID: PMC8870586 DOI: 10.3390/cancers14040911] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Alterations in glycosylation are frequently observed in cancer cells. Different strategies have been proposed to increase drug delivery to the tumor site in order to improve the therapeutic efficacy of anti-cancer drugs and avoid collateral cytotoxicity. The exploitation of drug delivery approaches directed to cancer-associated glycans has the potential to pave the way for better and more efficient personalized treatment practices. Such strategies taking advantage of aberrant cell surface glycosylation patterns enhance the targeting efficiency and optimize the delivery of clinically used drugs to cancer cells, with major potential for the clinical applications. Abstract Innovative strategies have been proposed to increase drug delivery to the tumor site and avoid cytotoxicity, improving the therapeutic efficacy of well-established anti-cancer drugs. Alterations in normal glycosylation processes are frequently observed in cancer cells and the resulting cell surface aberrant glycans can be used as direct molecular targets for drug delivery. In the present review, we address the development of strategies, such as monoclonal antibodies, antibody–drug conjugates and nanoparticles that specific and selectively target cancer-associated glycans in tumor cells. The use of nanoparticles for drug delivery encompasses novel applications in cancer therapy, including vaccines encapsulated in synthetic nanoparticles and specific nanoparticles that target glycoproteins or glycan-binding proteins. Here, we highlight their potential to enhance targeting approaches and to optimize the delivery of clinically approved drugs to the tumor microenvironment, paving the way for improved personalized treatment approaches with major potential importance for the pharmaceutical and clinical sectors.
Collapse
Affiliation(s)
- Francisca Diniz
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.D.); (P.C.); (H.O.D.); (B.S.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Pedro Coelho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.D.); (P.C.); (H.O.D.); (B.S.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Henrique O. Duarte
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.D.); (P.C.); (H.O.D.); (B.S.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.D.); (P.C.); (H.O.D.); (B.S.)
- INEB—Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- CESPU—Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, 4585-116 Gandra, Portugal
| | - Celso A. Reis
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.D.); (P.C.); (H.O.D.); (B.S.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Correspondence: (C.A.R.); (J.G.); Tel.: +351-220-408-800 (C.A.R. & J.G.)
| | - Joana Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.D.); (P.C.); (H.O.D.); (B.S.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- Correspondence: (C.A.R.); (J.G.); Tel.: +351-220-408-800 (C.A.R. & J.G.)
| |
Collapse
|
6
|
Hou L, Liu Y, Liu W, Balash M, Zhang H, Zhang Y, Zhang H, Zhang Z. In situ triggering antitumor efficacy of alcohol-abuse drug disulfiram through Cu-based metal-organic framework nanoparticles. Acta Pharm Sin B 2021; 11:2016-2030. [PMID: 34386335 PMCID: PMC8343114 DOI: 10.1016/j.apsb.2021.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/18/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
Although approved as an alcohol-abuse drug, disulfiram (DSF) exhibited potential anticancer activity when chelated with copper (Cu). However, the low level of intrinsic Cu, toxicity originated from exogenous Cu supplementation, and poor stability of DSF in vivo severely limited its application in cancer treatment. Herein, we proposed an in situ DSF antitumor efficacy triggered system, taking advantages of Cu-based metal-organic framework (MOF). In detail, DSF was encapsulated into Cu-MOF nanoparticles (NPs) during its formation, and the obtained NPs were coated with hyaluronic acid to enhance the tumor targetability and biocompatibility. Notably, DSF loaded Cu-MOF NPs maintained stability and integrity without Cu2+ leakage in blood circulation, thus showing excellent biosafety. Once accumulating at tumor site, NPs were internalized into tumor cells via receptor-mediated endocytosis and released DSF and Cu2+ simultaneously in the hyaluronidase-enriched and acidic intracellular tumor microenvironment. This profile lead to in situ chelation reaction between DSF and Cu2+, generating toxic DSF/Cu complex against tumor cells. Both in vitro and in vivo results demonstrated the programmed degradation and recombination property of Cu-based MOF NPs, which facilitated the tumor-specific chemotherapeutic effects of DSF. This system provided a promising strategy for the application of DSF in tumor therapy.
Collapse
Affiliation(s)
- Lin Hou
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
- School of Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Yanlong Liu
- School of Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Wei Liu
- School of Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Mervat Balash
- School of Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Hongling Zhang
- School of Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Huijuan Zhang
- School of Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| |
Collapse
|
7
|
Yusupov M, Privat-Maldonado A, Cordeiro RM, Verswyvel H, Shaw P, Razzokov J, Smits E, Bogaerts A. Oxidative damage to hyaluronan-CD44 interactions as an underlying mechanism of action of oxidative stress-inducing cancer therapy. Redox Biol 2021; 43:101968. [PMID: 33895486 PMCID: PMC8099558 DOI: 10.1016/j.redox.2021.101968] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/20/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Multiple cancer therapies nowadays rely on oxidative stress to damage cancer cells. Here we investigated the biological and molecular effect of oxidative stress on the interaction between CD44 and hyaluronan (HA), as interrupting their binding can hinder cancer progression. Our experiments demonstrated that the oxidation of HA decreased its recognition by CD44, which was further enhanced when both CD44 and HA were oxidized. The reduction of CD44-HA binding negatively affected the proliferative state of cancer cells. Our multi-level atomistic simulations revealed that the binding free energy of HA to CD44 decreased upon oxidation. The effect of HA and CD44 oxidation on CD44-HA binding was similar, but when both HA and CD44 were oxidized, the effect was much larger, in agreement with our experiments. Hence, our experiments and computations support our hypothesis on the role of oxidation in the disturbance of CD44-HA interaction, which can lead to the inhibition of proliferative signaling pathways inside the tumor cell to induce cell death.
Collapse
Affiliation(s)
- Maksudbek Yusupov
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium.
| | - Angela Privat-Maldonado
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium; Solid Tumor Immunology Group, Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium.
| | - Rodrigo M Cordeiro
- Centro de Ciências Naturais e Humanas, Universidade Federal Do ABC, Avenida Dos Estados 5001, CEP 09210-580, Santo André, SP, Brazil
| | - Hanne Verswyvel
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium; Solid Tumor Immunology Group, Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Priyanka Shaw
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium; Solid Tumor Immunology Group, Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Jamoliddin Razzokov
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium; Tashkent Institute of Irrigation and Agricultural Mechanization Engineers, Kori Niyoziy 39, 100000, Tashkent, Uzbekistan; Institute of Material Sciences, Uzbek Academy of Sciences, Chingiz Aytmatov 2b, 100084, Tashkent, Uzbekistan
| | - Evelien Smits
- Solid Tumor Immunology Group, Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Annemie Bogaerts
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium.
| |
Collapse
|
8
|
Navarro-Marchal SA, Griñán-Lisón C, Entrena JM, Ruiz-Alcalá G, Tristán-Manzano M, Martin F, Pérez-Victoria I, Peula-García JM, Marchal JA. Anti-CD44-Conjugated Olive Oil Liquid Nanocapsules for Targeting Pancreatic Cancer Stem Cells. Biomacromolecules 2021; 22:1374-1388. [PMID: 33724003 DOI: 10.1021/acs.biomac.0c01546] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The latest trends in cancer research and nanomedicine focus on using nanocarriers to target cancer stem cells (CSCs). Specifically, lipid liquid nanocapsules are usually developed as nanocarriers for lipophilic drug delivery. Here, we developed olive oil liquid NCs (O2LNCs) functionalized by covalent coupling of an anti-CD44-fluorescein isothiocyanate antibody (αCD44). First, O2LNCs are formed by a core of olive oil surrounded by a shell containing phospholipids, a nonionic surfactant, and deoxycholic acid molecules. Then, O2LNCs were coated with an αCD44 antibody (αCD44-O2LNC). The optimization of an αCD44 coating procedure, a complete physicochemical characterization, as well as clear evidence of their efficacy in vitro and in vivo were demonstrated. Our results indicate the high targeted uptake of these αCD44-O2LNCs, and the increased antitumor efficacy (up to four times) of paclitaxel-loaded-αCD44-O2LNC compared to free paclitaxel in pancreatic CSCs (PCSCs). Also, αCD44-O2LNCs were able to selectively target PCSCs in an orthotopic xenotransplant in vivo model.
Collapse
Affiliation(s)
- Saúl A Navarro-Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18071 Granada, Spain.,Department of Applied Physics, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18071 Granada, Spain
| | - José-Manuel Entrena
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, Armilla, 18100 Granada, Spain.,Animal Behavior Research Unit, Scientific Instrumentation Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, Armilla, 18100 Granada, Spain
| | - Gloria Ruiz-Alcalá
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18071 Granada, Spain
| | - María Tristán-Manzano
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Francisco Martin
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Ignacio Pérez-Victoria
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Parque Tecnológico de Ciencias de la Salud, Avda. del Conocimiento 34, Armilla, 18016 Granada, Spain
| | - José Manuel Peula-García
- Biocolloids and Fluids Physics Group, Faculty of Sciences, University of Granada, 18014 Granada, Spain.,Department of Applied Physics II, University of Málaga, 29071 Málaga, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18071 Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| |
Collapse
|
9
|
Yoshikawa C, Nakaji-Hirabayashi T, Nishijima N, Nonsuwan P, Toh RJ, Kowalczyk W, Thissen H. Ultra-low fouling photocrosslinked coatings for the selective capture of cells expressing CD44. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111630. [PMID: 33545815 DOI: 10.1016/j.msec.2020.111630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/27/2020] [Accepted: 10/13/2020] [Indexed: 11/27/2022]
Abstract
The effective control of biointerfacial interactions is of outstanding interest in a broad range of biomedical applications, ranging from cell culture tools to biosensors and implantable medical devices. For many of these applications, highly specific interactions between cells and material surfaces are desired. Sophisticated control over these interactions requires reducing or preventing non-specific interactions on the one hand and displaying highly specific signals that can be recognized by extracellular receptors on the other. We have recently developed ultra-low fouling coatings that can be applied in a single step using photoreactive copolymers of 2-hydroxypropyl acrylamide and N-benzophenone acrylamide. Here, we have expanded this approach by incorporating polymerizable peptide monomers into these copolymers. The monomers QQGWFGAGK(acrylamide) and acrylamide-GAGQQGWF were synthesized after identifying the QQGWF sequence as a binding motif for CD44 by phage display for the first time. Our results demonstrate that UV-crosslinked coatings fabricated using the QQGWFGAGK(acrylamide) monomer are effective at selectively binding hMSC in the presence of HepG2 and HEK293 cells due to the difference in CD44 expression. Our results also demonstrate that the peptide modified coatings retain their low biofouling character using a BCA protein binding assay as well as an E. coli bacterial attachment assay over a 24 h period. Our approach provides an alternative to traditional integrin-mediated selective cell binding on surfaces and opens the door to new diagnostic applications, exploiting the fact that the transmembrane protein CD44 is highly expressed in multiple diseases.
Collapse
Affiliation(s)
- Chiaki Yoshikawa
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-2-1, Tsukuba, Ibaraki 305-0047, Japan.
| | - Tadashi Nakaji-Hirabayashi
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-2-1, Tsukuba, Ibaraki 305-0047, Japan; Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama, Toyama 930-8555, Japan; Graduate School of Innovative Life Science, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan.
| | - Nanami Nishijima
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-2-1, Tsukuba, Ibaraki 305-0047, Japan; Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama, Toyama 930-8555, Japan
| | - Punnida Nonsuwan
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-2-1, Tsukuba, Ibaraki 305-0047, Japan
| | - Rou Jun Toh
- CSIRO Manufacturing, Research Way, Clayton, Victoria 3168, Australia
| | - Wioleta Kowalczyk
- CSIRO Manufacturing, Research Way, Clayton, Victoria 3168, Australia
| | - Helmut Thissen
- CSIRO Manufacturing, Research Way, Clayton, Victoria 3168, Australia.
| |
Collapse
|
10
|
Choi KY, Han HS, Lee ES, Shin JM, Almquist BD, Lee DS, Park JH. Hyaluronic Acid-Based Activatable Nanomaterials for Stimuli-Responsive Imaging and Therapeutics: Beyond CD44-Mediated Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1803549. [PMID: 30773699 DOI: 10.1002/adma.201803549] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 12/27/2018] [Indexed: 05/24/2023]
Abstract
There is a rapidly increasing interest in developing stimuli-responsive nanomaterials for treating a variety of diseases. By enabling the activation of function locally at the sites of interest, it is possible to increase therapeutic efficacy significantly while simultaneously reducing adverse side effects. While there are many sophisticated nanomaterials available, they are often highly complex and not easily transferrable to industrial scales and clinical settings. However, nanomaterials based on hyaluronic acid offer a compelling strategy for reducing their complexity while retaining several desirable benefits such as active targeting and stimuli-responsive degradation. Herein, the basic properties of hyaluronic acid, its binding partners, and natural routes for degradation by hyaluronidases-hyaluronic-acid-degrading enzymes-and oxidative stresses are discussed. Recent advances in designing hyaluronic acid-based, actively targeted, hyaluronidase- or reactive-oxygen-species-responsive nanomaterials for both diagnostic imaging and therapeutic delivery, which go beyond merely the classical targeting of CD44, are summarized.
Collapse
Affiliation(s)
- Ki Young Choi
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea
| | - Hwa Seung Han
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea
| | - Eun Sook Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jung Min Shin
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | | | - Doo Sung Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
11
|
Liang H, Zhou Z, Luo R, Sang M, Liu B, Sun M, Qu W, Feng F, Liu W. Tumor-specific activated photodynamic therapy with an oxidation-regulated strategy for enhancing anti-tumor efficacy. Am J Cancer Res 2018; 8:5059-5071. [PMID: 30429886 PMCID: PMC6217056 DOI: 10.7150/thno.28344] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022] Open
Abstract
Photodynamic therapy relies on photosensitizers to generate cytotoxic reactive oxygen species (ROS) resulting in the apoptois of tumor cells. However, there is an antioxidant system that impedes the elevation of oxidation levels in tumor cells. Thus, photodynamic therapy may exhibit insufficient curative effects due to ungenerous reactive oxygen species levels. Herein, we describe tumor-specific activated photodynamic therapy using an oxidation-regulating strategy. Methods: We first synthesised a reactive oxygen species-sensitive amphipathic prodrug of gambogic acid-grafted hyaluronic acid (HA-GA). The hydrophobic photosensitizer chlorin e6 (Ce6) was then loaded into HA-GA by hydrophobic interactions between GA and Ce6, forming amphipathic nanomicelles (HA-GA@Ce6). The ROS-responsive behavior, cytotoxicity, cell uptake, tumor cell killing, in vivo biodistribution and in vivo anti-tumor efficacy of HA-GA@Ce6 were investigated. The in vitro and in vivo experiments were performed on 4T1 murine breast cancer cells and 4T1 tumor model. Results: We validated that the micelles of HA-GA@Ce6 showed stronger cell uptake in 4T1 tumor cells and lower cytotoxicity in normal cells compared with free Ce6 and GA, which exhibited the benefits of nanomicelles on enhancing the tumor cell acumulation and reducing the side effects on normal cells synchronously. Additionally, the cytotoxic free radicals of photodynamic therapy were generated after irradiation and the high oxidation levels activated the ROS-sensitive GA prodrug efficiently, which killed the tumor cells and depleted intracellular glutathione (GSH), thereby impairing antioxidant levels and enhancing photodynamic therapy. Conclusion: With the successfully eradicated tumor growth in vivo. Our work represents a new photodynamic therapy concept, achieving superior anti-tumor efficacy by reducing intracellular antioxidant levels.
Collapse
|
12
|
Karamanos NK, Piperigkou Z, Theocharis AD, Watanabe H, Franchi M, Baud S, Brézillon S, Götte M, Passi A, Vigetti D, Ricard-Blum S, Sanderson RD, Neill T, Iozzo RV. Proteoglycan Chemical Diversity Drives Multifunctional Cell Regulation and Therapeutics. Chem Rev 2018; 118:9152-9232. [PMID: 30204432 DOI: 10.1021/acs.chemrev.8b00354] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi 480-1195, Japan
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini 47100, Italy
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Laboratoire SiRMa, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster 48149, Germany
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, CNRS, UMR 5246, Institute of Molecular and Supramolecular Chemistry and Biochemistry, Villeurbanne 69622, France
| | - Ralph D. Sanderson
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| |
Collapse
|
13
|
Su X, Xue Y, Wei J, Huo X, Gong Y, Zhang H, Han R, Chen Y, Chen H, Chen J. Establishment and Characterization of gc-006-03, a Novel Human Signet Ring Cell Gastric Cancer Cell Line Derived from Metastatic Ascites. J Cancer 2018; 9:3236-3246. [PMID: 30271482 PMCID: PMC6160672 DOI: 10.7150/jca.26051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022] Open
Abstract
Signet ring cell gastric cancer (SRCGC) is a special type of gastric cancer with rapid progression and poor prognosis. However, few available SRCGC cell lines from Chinese patients can be used for research, the molecular mechanism of its growth and metastasis is still incompletely understood. In this study, we established and characterized a novel SRCGC cell line, gc-006-03.The cells showed a tendency to pile up without contact inhibition. G-band karyotypes of gc-006-03 were revealed hypotriploid with a modal chromosome number of 51. Immunohistochemistry analysis showed that the cells were positive for CEA, CK7, CDX2 and Ki-67(45%), and negative for CK20, TTF1and Li-cadherin. Flow cytometry analysis showed that gc-006-3 had 25% of CD44+ cells. The cells possessed strong clonality and high plating efficiency, and the doubling time was 36h. The cells grew vigorously for more than 100 passages in serial culture. Meanwhile, the cells showed a high rate of tumor formation. Tumors were observed in all of the nude mice (5/5) given injections of the cells. The metastatic capability of the cell line was found in zebrafish injected the cells. The results of whole genome sequencing revealed the unique genomic characteristics of gc-006-03. In summary, this new stable cell line may be useful in basic and clinical research on gastric signet ring cell carcinoma.
Collapse
Affiliation(s)
- Xinyu Su
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Radiation Oncology, the Affiliated Huai'an Hospital of Xuzhou Medical College, Huai'an, China
| | - Yiqi Xue
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jingsun Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xinying Huo
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yang Gong
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Honghong Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Rongbo Han
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuetong Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hong Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jinfei Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Saneja A, Arora D, Kumar R, Dubey RD, Panda AK, Gupta PN. CD44 targeted PLGA nanomedicines for cancer chemotherapy. Eur J Pharm Sci 2018; 121:47-58. [PMID: 29777858 DOI: 10.1016/j.ejps.2018.05.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/19/2018] [Accepted: 05/14/2018] [Indexed: 12/28/2022]
Abstract
In recent years scientific community has drawn a great deal of attention towards understanding the enigma of cluster of differentiation-44 (CD44) in order to deliver therapeutic agents more selectively towards tumor tissues. Moreover, its over-expression in variety of solid tumors has attracted drug delivery researchers to target this receptor with nanomedicines. Conventional nanomedicines based on biodegradable polymers such as poly(lactide-co-glycolide) (PLGA) are often associated with insufficient cellular uptake by cancer cells, due to lack of active targeting moiety on their surface. Therefore, to address this limitation, CD44 targeted PLGA nanomedicines has gained considerable interest for enhancing the efficacy of chemotherapeutic agents. In this review, we have elaborately discussed the recent progress in the design and synthesis of CD44 targeted PLGA nanomedicines used to improve tumor-targeted drug delivery. We have also discussed strategies based on co-targeting of CD44 with other targeting moieties such as folic acid, human epidermal growth factor 2 (HER2), monoclonal antibodies using PLGA based nanomedicines.
Collapse
Affiliation(s)
- Ankit Saneja
- Product Development Cell-II, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India; Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| | - Divya Arora
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Robin Kumar
- Product Development Cell-II, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Ravindra Dhar Dubey
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Amulya K Panda
- Product Development Cell-II, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Prem N Gupta
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| |
Collapse
|
15
|
Piperigkou Z, Götte M, Theocharis AD, Karamanos NK. Insights into the key roles of epigenetics in matrix macromolecules-associated wound healing. Adv Drug Deliv Rev 2018; 129:16-36. [PMID: 29079535 DOI: 10.1016/j.addr.2017.10.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/14/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023]
Abstract
Extracellular matrix (ECM) is a dynamic network of macromolecules, playing a regulatory role in cell functions, tissue regeneration and remodeling. Wound healing is a tissue repair process necessary for the maintenance of the functionality of tissues and organs. This highly orchestrated process is divided into four temporally overlapping phases, including hemostasis, inflammation, proliferation and tissue remodeling. The dynamic interplay between ECM and resident cells exerts its critical role in many aspects of wound healing, including cell proliferation, migration, differentiation, survival, matrix degradation and biosynthesis. Several epigenetic regulatory factors, such as the endogenous non-coding microRNAs (miRNAs), are the drivers of the wound healing response. microRNAs have pivotal roles in regulating ECM composition during wound healing and dermal regeneration. Their expression is associated with the distinct phases of wound healing and they serve as target biomarkers and targets for systematic regulation of wound repair. In this article we critically present the importance of epigenetics with particular emphasis on miRNAs regulating ECM components (i.e. glycoproteins, proteoglycans and matrix proteases) that are key players in wound healing. The clinical relevance of miRNA targeting as well as the delivery strategies designed for clinical applications are also presented and discussed.
Collapse
|
16
|
|
17
|
Zhou Z, Li H, Wang K, Guo Q, Li C, Jiang H, Hu Y, Oupicky D, Sun M. Bioreducible Cross-Linked Hyaluronic Acid/Calcium Phosphate Hybrid Nanoparticles for Specific Delivery of siRNA in Melanoma Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:14576-14589. [PMID: 28393529 DOI: 10.1021/acsami.6b15347] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
This study introduces a novel cross-linking strategy capable of successfully stabilizing CaP nanoparticles and stimuli-responsive small interfering RNA (siRNA) release. We synthesized a polysaccharide derivative thiolated hyaluronic acid (HA-SH), which was slightly modified but multifunctional and developed a smart redox-responsive delivery system. siRNA was efficaciously condensed by calcium phosphate (CaP) via electrostatic interaction to form a positively charged inner "core". Disulfide cross-linked HA (HA-ss-HA) was formed and played a role as an anionic outer "shell" to stabilize the CaP core. We demonstrated that the nanoparticles were stable both in the storage milieu and systemic circulation, thus overcoming the most serious disadvantage of CaP nanoparticles for gene delivery. Meanwhile, this smart system could selectively release siRNA into the cytosol by both a GSH-triggered disassembly and successful endosomal escape. Therefore, the hybrid delivery system achieved an 80% gene-silencing efficiency in vitro for both luciferase and Bcl2. Silencing of Bcl2 resulted in dramatic apoptosis of B16F10 cells. Besides, equipped with the tumor-targeting component HA, the nanoparticles significantly suppressed the growth of B16F10 xenograft tumor in mice. The anionic HA-ss-HA-equipped nanoparticles showed no apparent toxicity in vitro or in vivo, as well as showed a high transfection efficiency. Taken together, this redox-responsive, tumor-targeting smart anionic nanoparticle holds great promise for exploitation in functionalized siRNA delivery and tumor therapy.
Collapse
Affiliation(s)
- Zhanwei Zhou
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University , Nanjing 210009, China
| | - Huipeng Li
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University , Nanjing 210009, China
| | - Kaikai Wang
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University , Nanjing 210009, China
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University , Nanjing 210093, China
| | - Qian Guo
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University , Nanjing 210009, China
| | - Chenzi Li
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University , Nanjing 210009, China
| | - Hulin Jiang
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University , Nanjing 210009, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University , Nanjing 210093, China
| | - David Oupicky
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University , Nanjing 210009, China
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Minjie Sun
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University , Nanjing 210009, China
| |
Collapse
|
18
|
Abstract
With the refinement of functional properties, the interest around biodegradable materials, in biorelated applications and, in particular, in their use as controlled drug-delivery systems, increased in the last decades. Biodegradable materials are an ideal platform to obtain nanoparticles for spatiotemporal controlled drug delivery for the in vivo administration, thanks to their biocompatibility, functionalizability, the control exerted on delivery rates and the complete degradation. Their application in systems for cancer treatment, brain and cardiovascular diseases is already a consolidated practice in research, while the bench-to-bedside translation is still late. This review aims at summarizing reported applications of biodegradable materials to obtain drug-delivery nanoparticles in the last few years, giving a complete overview of pros and cons related to degradable nanomedicaments.
Collapse
|
19
|
Roles and targeting of the HAS/hyaluronan/CD44 molecular system in cancer. Matrix Biol 2016; 59:3-22. [PMID: 27746219 DOI: 10.1016/j.matbio.2016.10.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 02/07/2023]
Abstract
Synthesis, deposition, and interactions of hyaluronan (HA) with its cellular receptor CD44 are crucial events that regulate the onset and progression of tumors. The intracellular signaling pathways initiated by HA interactions with CD44 leading to tumorigenic responses are complex. Moreover, HA molecules may perform dual functions depending on their concentration and size. Overexpression of variant isoforms of CD44 (CD44v) is most commonly linked to cancer progression, whereas their loss is associated with inhibition of tumor growth. In this review, we highlight that the regulation of HA synthases (HASes) by post-translational modifications, such as O-GlcNAcylation and ubiquitination, environmental factors and the action of microRNAs is important for HA synthesis and secretion in the tumor microenvironment. Moreover, we focus on the roles and interactions of CD44 with various proteins that reside extra- and intracellularly, as well as on cellular membranes with particular reference to the CD44-HA axis in cancer stem cell functions, and the importance of CD44/CD44v6 targeting to inhibit tumorigenesis.
Collapse
|
20
|
Nascimento TL, Hillaireau H, Vergnaud J, Fattal E. Lipid-based nanosystems for CD44 targeting in cancer treatment: recent significant advances, ongoing challenges and unmet needs. Nanomedicine (Lond) 2016; 11:1865-87. [DOI: 10.2217/nnm-2016-5000] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Extensive experimental evidence demonstrates the important role of hyaluronic acid (HA)-CD44 interaction in cell proliferation and migration, inflammation and tumor growth. Taking advantage of this interaction, the design of HA-modified nanocarriers has been investigated for targeting CD44-overexpressing cells with the purpose of delivering drugs to cancer or inflammatory cells. The effect of such modification on targeting efficacy is influenced by several factors. In this review, we focus on the impact of HA-modification on the characteristics of lipid-based nanoparticles. We try to understand how these modifications influence particle physicochemical properties, interaction with CD44 receptors, intracellular trafficking pathways, toxicity, complement/macrophage activation and pharmacokinetics. Our aim is to provide insight in tailoring particle modification by HA in order to design more efficient CD44-targeting lipid nanocarriers.
Collapse
Affiliation(s)
- Thais Leite Nascimento
- Institut Galien Paris-Sud, Faculté de pharmacie, Université Paris-Sud, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CAPES Foundation, Ministry of Education of Brazil, Brasília – DF 70040-020, Brazil
| | - Hervé Hillaireau
- Institut Galien Paris-Sud, Faculté de pharmacie, Université Paris-Sud, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| | - Juliette Vergnaud
- Institut Galien Paris-Sud, Faculté de pharmacie, Université Paris-Sud, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| | - Elias Fattal
- Institut Galien Paris-Sud, Faculté de pharmacie, Université Paris-Sud, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| |
Collapse
|
21
|
Piperigkou Z, Karamanou K, Engin AB, Gialeli C, Docea AO, Vynios DH, Pavão MS, Golokhvast KS, Shtilman MI, Argiris A, Shishatskaya E, Tsatsakis AM. Emerging aspects of nanotoxicology in health and disease: From agriculture and food sector to cancer therapeutics. Food Chem Toxicol 2016; 91:42-57. [DOI: 10.1016/j.fct.2016.03.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 02/07/2023]
|
22
|
Wang G, Zhang F, Tian R, Zhang L, Fu G, Yang L, Zhu L. Nanotubes-Embedded Indocyanine Green-Hyaluronic Acid Nanoparticles for Photoacoustic-Imaging-Guided Phototherapy. ACS APPLIED MATERIALS & INTERFACES 2016; 8:5608-17. [PMID: 26860184 PMCID: PMC4930365 DOI: 10.1021/acsami.5b12400] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Phototherapy is a light-triggered treatment for tumor ablation and growth inhibition via photodynamic therapy (PDT) and photothermal therapy (PTT). Despite extensive studies in this area, a major challenge is the lack of selective and effective phototherapy agents that can specifically accumulate in tumors to reach a therapeutic concentration. Although recent attempts have produced photosensitizers complexed with photothermal nanomaterials, the tedious preparation steps and poor tumor efficiency of therapy still hampers the broad utilization of these nanocarriers. Herein, we developed a CD44 targeted photoacoustic (PA) nanophototherapy agent by conjugating Indocyanine Green (ICG) to hyaluronic acid nanoparticles (HANPs) encapsulated with single-walled carbon nanotubes (SWCNTs), resulting in a theranostic nanocomplex of ICG-HANP/SWCNTs (IHANPT). We fully characterized its physical features as well as PA imaging and photothermal and photodynamic therapy properties in vitro and in vivo. Systemic delivery of IHANPT theranostic nanoparticles led to the accumulation of the targeted nanoparticles in tumors in a human cancer xenograft model in nude mice. PA imaging confirmed targeted delivery of the IHANPT nanoparticles into tumors (T/M ratio = 5.19 ± 0.3). The effect of phototherapy was demonstrated by low-power laser irradiation (808 nm, 0.8 W/cm(2)) to induce efficient photodynamic effect from ICG dye. The photothermal effect from the ICG and SWCNTs rapidly raised the tumor temperature to 55.4 ± 1.8 °C. As the result, significant tumor growth inhibition and marked induction of tumor cell death and necrosis were observed in the tumors in the tumors. There were no apparent systemic and local toxic effects found in the mice. The dynamic thermal stability of IHANPT was studied to ensure that PTT does not affect ICG-dependent PDT in phototherapy. Therefore, our results highlight imaging property and therapeutic effect of the novel IHANPT theranostic nanoparticle for CD44 targeted and PA image-guided dual PTT and PDT cancer therapy.
Collapse
Affiliation(s)
- Guohao Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361005, China
| | - Fan Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361005, China
| | - Rui Tian
- Department of Ophthalmology Second Hospital, Jilin University, Changchun, Jilin 130033, China
- Corresponding Authors: (R. Tian). Tel.: (+)86-592-2880642. Fax: (+)86-592-2880642. (L. Zhu)
| | - Liwen Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361005, China
| | - Guifeng Fu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361005, China
| | - Lily Yang
- Departments of Surgery and Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Lei Zhu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361005, China
- Departments of Surgery and Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Corresponding Authors: (R. Tian). Tel.: (+)86-592-2880642. Fax: (+)86-592-2880642. (L. Zhu)
| |
Collapse
|
23
|
Kamaly N, Yameen B, Wu J, Farokhzad OC. Degradable Controlled-Release Polymers and Polymeric Nanoparticles: Mechanisms of Controlling Drug Release. Chem Rev 2016; 116:2602-63. [PMID: 26854975 PMCID: PMC5509216 DOI: 10.1021/acs.chemrev.5b00346] [Citation(s) in RCA: 1625] [Impact Index Per Article: 180.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nazila Kamaly
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Basit Yameen
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jun Wu
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Omid C. Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
24
|
Dosio F, Arpicco S, Stella B, Fattal E. Hyaluronic acid for anticancer drug and nucleic acid delivery. Adv Drug Deliv Rev 2016; 97:204-36. [PMID: 26592477 DOI: 10.1016/j.addr.2015.11.011] [Citation(s) in RCA: 413] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 01/06/2023]
Abstract
Hyaluronic acid (HA) is widely used in anticancer drug delivery, since it is biocompatible, biodegradable, non-toxic, and non-immunogenic; moreover, HA receptors are overexpressed on many tumor cells. Exploiting this ligand-receptor interaction, the use of HA is now a rapidly-growing platform for targeting CD44-overexpressing cells, to improve anticancer therapies. The rationale underlying approaches, chemical strategies, and recent advances in the use of HA to design drug carriers for delivering anticancer agents, are reviewed. Comprehensive descriptions are given of HA-based drug conjugates, particulate carriers (micelles, liposomes, nanoparticles, microparticles), inorganic nanostructures, and hydrogels, with particular emphasis on reports of preclinical/clinical results.
Collapse
|
25
|
Yan Y, Zuo X, Wei D. Concise Review: Emerging Role of CD44 in Cancer Stem Cells: A Promising Biomarker and Therapeutic Target. Stem Cells Transl Med 2015; 4:1033-43. [PMID: 26136504 DOI: 10.5966/sctm.2015-0048] [Citation(s) in RCA: 456] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/26/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED The reception and integration of the plethora of signals a cell receives from its microenvironment determines the cell's fate. CD44 functions as a receptor for hyaluronan and many other extracellular matrix components, as well as a cofactor for growth factors and cytokines, and thus, CD44 is a signaling platform that integrates cellular microenvironmental cues with growth factor and cytokine signals and transduces signals to membrane-associated cytoskeletal proteins or to the nucleus to regulate a variety of gene expression levels related to cell-matrix adhesion, cell migration, proliferation, differentiation, and survival. Accumulating evidence indicates that CD44, especially CD44v isoforms, are cancer stem cell (CSC) markers and critical players in regulating the properties of CSCs, including self-renewal, tumor initiation, metastasis, and chemoradioresistance. Furthermore, there is ample evidence that CD44, especially CD44v isoforms, are valuable prognostic markers in various types of tumors. Therefore, therapies that target CD44 may destroy the CSC population, and this holds great promise for the cure of life-threatening cancers. However, many challenges remain to determining how best to use CD44 as a biomarker and therapeutic target. Here we summarize the current findings concerning the critical role of CD44/CD44v in the regulation of cancer stemness and the research status of CD44/CD44v as biomarkers and therapeutic targets in cancer. We also discuss the current challenges and future directions that may lead to the best use of CD44/CD44v for clinical applications. SIGNIFICANCE Mounting evidence indicates that cancer stem cells (CSCs) are mainly responsible for cancer aggressiveness, drug resistance, and tumor relapse. CD44, especially CD44v isoforms, have been identified as CSC surface markers for isolating and enriching CSCs in different types of cancers. The current findings concerning the critical role of CD44/CD44v in regulation of cancer stemness and the research status of CD44/CD44v as biomarkers and therapeutic targets in cancer are summarized. The current challenges and future directions that may lead to best use of CD44/CD44v for clinical applications are also discussed.
Collapse
Affiliation(s)
- Yongmin Yan
- Departments of Gastroenterology, Hepatology & Nutrition and Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; School of Medical Sciences and Laboratory Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Xiangsheng Zuo
- Departments of Gastroenterology, Hepatology & Nutrition and Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; School of Medical Sciences and Laboratory Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Daoyan Wei
- Departments of Gastroenterology, Hepatology & Nutrition and Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; School of Medical Sciences and Laboratory Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| |
Collapse
|
26
|
Zöller M. CD44, Hyaluronan, the Hematopoietic Stem Cell, and Leukemia-Initiating Cells. Front Immunol 2015; 6:235. [PMID: 26074915 PMCID: PMC4443741 DOI: 10.3389/fimmu.2015.00235] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/30/2015] [Indexed: 12/14/2022] Open
Abstract
CD44 is an adhesion molecule that varies in size due to glycosylation and insertion of so-called variant exon products. The CD44 standard isoform (CD44s) is highly expressed in many cells and most abundantly in cells of the hematopoietic system, whereas expression of CD44 variant isoforms (CD44v) is more restricted. CD44s and CD44v are known as stem cell markers, first described for hematopoietic stem cells and later on confirmed for cancer- and leukemia-initiating cells. Importantly, both abundantly expressed CD44s as well as CD44v actively contribute to the maintenance of stem cell features, like generating and embedding in a niche, homing into the niche, maintenance of quiescence, and relative apoptosis resistance. This is surprising, as CD44 is not a master stem cell gene. I here will discuss that the functional contribution of CD44 relies on its particular communication skills with neighboring molecules, adjacent cells and, last not least, the surrounding matrix. In fact, it is the interaction of the hyaluronan receptor CD44 with its prime ligand, which strongly assists stem cells to fulfill their special and demanding tasks. Recent fundamental progress in support of this “old” hypothesis, which may soon pave the way for most promising new therapeutics, is presented for both hematopoietic stem cell and leukemia-initiating cell. The contribution of CD44 to the generation of a stem cell niche, to homing of stem cells in their niche, to stem cell quiescence and apoptosis resistance will be in focus.
Collapse
Affiliation(s)
- Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery , Heidelberg , Germany
| |
Collapse
|
27
|
Adjei IM, Blanka S. Modulation of the tumor microenvironment for cancer treatment: a biomaterials approach. J Funct Biomater 2015; 6:81-103. [PMID: 25695337 PMCID: PMC4384103 DOI: 10.3390/jfb6010081] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/07/2014] [Accepted: 02/12/2015] [Indexed: 12/26/2022] Open
Abstract
Tumors are complex tissues that consist of stromal cells, such as fibroblasts, immune cells and mesenchymal stem cells, as well as non-cellular components, in addition to neoplastic cells. Increasingly, there is evidence to suggest that these non-neoplastic cell components support cancer initiation, progression and metastasis and that their ablation or reprogramming can inhibit tumor growth. Our understanding of the activities of different parts of the tumor stroma in advancing cancer has been improved by the use of scaffold and matrix-based 3D systems originally developed for regenerative medicine. Additionally, drug delivery systems made from synthetic and natural biomaterials deliver drugs to kill stromal cells or reprogram the microenvironment for tumor inhibition. In this article, we review the impact of 3D tumor models in increasing our understanding of tumorigenesis. We also discuss how different drug delivery systems aid in the reprogramming of tumor stroma for cancer treatment.
Collapse
Affiliation(s)
- Isaac M Adjei
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| | - Sharma Blanka
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
28
|
Lu KY, Lin CW, Hsu CH, Ho YC, Chuang EY, Sung HW, Mi FL. FRET-based dual-emission and pH-responsive nanocarriers for enhanced delivery of protein across intestinal epithelial cell barrier. ACS APPLIED MATERIALS & INTERFACES 2014; 6:18275-18289. [PMID: 25260022 DOI: 10.1021/am505441p] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The oral route is a convenient and commonly employed way for drug delivery. However, therapeutic proteins have poor bioavailability upon oral administration due to the impermeable barrier from intestinal epithelial tight junction (TJ). Moreover, the pH of the small intestine varies among different regions of the intestinal tract where digestion and absorption occur at different levels. In this study, a tunable dual-emitting and pH-responsive nanocarrier that can alter the fluorescent color and emission intensity in response to pH changes and can trigger the opening of intestinal epithelial TJ at different levels were developed from chitosan-N-arginine and poly(γ-glutamic acid)-taurine conjugates. As pH increased from 6.0 to 8.0, the binding affinity of the oppositely charged polyions decreased, whereas the ratio of the intensity of the donor-to-acceptor emission intensity (ID/IA) increased by 27-fold. The fluorescent and pH-responsive nanocarrier was able to monitor the pH change of intestinal environment and to control the release of an anti-angiogenic protein in response to the pH gradient. The nanocarrier triggered the opening of intestinal epithelial TJ and consequently enhanced the permeation of the released protein through the intestinal epithelial barrier model (Caco-2 cell monolayer) to inhibit tube formation of human umbilical vein endothelial cells.
Collapse
Affiliation(s)
- Kun-Ying Lu
- Institute of Organic and Polymeric Materials, National Taipei University of Technology , Taipei 10608, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|