1
|
Hosseini-Kharat M, Bremmell KE, Grubor-Bauk B, Prestidge CA. Enhancing non-viral DNA delivery systems: Recent advances in improving efficiency and target specificity. J Control Release 2025; 378:170-194. [PMID: 39647508 DOI: 10.1016/j.jconrel.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
DNA-based therapies are often limited by challenges such as stability, long-term integration, low transfection efficiency, and insufficient targeted DNA delivery. This review focuses on recent progress in the design of non-viral delivery systems for enhancing targeted DNA delivery and modulation of therapeutic efficiency. Cellular uptake and intracellular trafficking mechanisms play a crucial role in optimizing gene delivery efficiency. There are two main strategies employed to improve the efficiency of gene delivery vectors: (i) explore different administration routes (e.g., mucosal, intravenous, intramuscular, subcutaneous, intradermal, intratumoural, and intraocular) that best facilitates optimal uptake into the targeted cells and organs and (ii) modify the delivery vectors with cell-specific ligands (e.g., natural ligands, antibodies, peptides, carbohydrates, or aptamers) that enable targeted uptake to specific cells with higher specificity and improved biodistribution. We describe how recent progress in employing these DNA delivery strategies is advancing the field and increasing the clinical translation and ultimate clinical application of DNA therapies.
Collapse
Affiliation(s)
- Mahboubeh Hosseini-Kharat
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia
| | - Kristen E Bremmell
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia
| | - Branka Grubor-Bauk
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Clive A Prestidge
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
2
|
Basu A, Namporn T, Ruenraroengsak P. Critical Review in Designing Plant-Based Anticancer Nanoparticles against Hepatocellular Carcinoma. Pharmaceutics 2023; 15:1611. [PMID: 37376061 DOI: 10.3390/pharmaceutics15061611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC), accounting for 85% of liver cancer cases, continues to be the third leading cause of cancer-related deaths worldwide. Although various forms of chemotherapy and immunotherapy have been investigated in clinics, patients continue to suffer from high toxicity and undesirable side effects. Medicinal plants contain novel critical bioactives that can target multimodal oncogenic pathways; however, their clinical translation is often challenged due to poor aqueous solubility, low cellular uptake, and poor bioavailability. Nanoparticle-based drug delivery presents great opportunities in HCC therapy by increasing selectivity and transferring sufficient doses of bioactives to tumor areas with minimal damage to adjacent healthy cells. In fact, many phytochemicals encapsulated in FDA-approved nanocarriers have demonstrated the ability to modulate the tumor microenvironment. In this review, information about the mechanisms of promising plant bioactives against HCC is discussed and compared. Their benefits and risks as future nanotherapeutics are underscored. Nanocarriers that have been employed to encapsulate both pure bioactives and crude extracts for application in various HCC models are examined and compared. Finally, the current limitations in nanocarrier design, challenges related to the HCC microenvironment, and future opportunities are also discussed for the clinical translation of plant-based nanomedicines from bench to bedside.
Collapse
Affiliation(s)
- Aalok Basu
- Department of Pharmacy, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Rd., Rajathevi, Bangkok 10400, Thailand
| | - Thanaphon Namporn
- Department of Pharmacy, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Rd., Rajathevi, Bangkok 10400, Thailand
| | - Pakatip Ruenraroengsak
- Department of Pharmacy, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Rd., Rajathevi, Bangkok 10400, Thailand
| |
Collapse
|
3
|
Diaz-Dussan D, Peng YY, Rashed FB, Macdonald D, Weinfeld M, Kumar P, Narain R. Optimized Carbohydrate-Based Nanogel Formulation to Sensitize Hypoxic Tumors. Mol Pharm 2023. [PMID: 37148327 DOI: 10.1021/acs.molpharmaceut.3c00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Solid tumors are often poorly vascularized, which impairs oxygen supply and drug delivery to the cells. This often leads to genetic and translational adaptations that promote tumor progression, invasion, metastasis, and resistance to conventional chemo-/radiotherapy and immunotherapy. A hypoxia-directed nanosensitizer formulation of a hypoxia-activated prodrug (HAP) was developed by encapsulating iodoazomycin arabinofuranoside (IAZA), a 2-nitroimidazole nucleoside-based HAP, in a functionally modified carbohydrate-based nanogel, facilitating delivery and accrual selectively in the hypoxic head and neck and prostate cancer cells. Although IAZA has been reported as a clinically validated hypoxia diagnostic agent, recent studies have pointed to its promising hypoxia-selective anti-tumor properties, which make IAZA an excellent candidate for further exploration as a multimodal theranostic of hypoxic tumors. The nanogels are composed of a galactose-based shell with an inner core of thermoresponsive (di(ethylene glycol) methyl ethyl methacrylate) (DEGMA). Optimization of the nanogels led to high IAZA-loading capacity (≅80-88%) and a slow time-controlled release over 50 h. Furthermore, nanoIAZA (encapsulated IAZA) displayed superior in vitro hypoxia-selective cytotoxicity and radiosensitization in comparison to free IAZA in the head and neck (FaDu) and prostate (PC3) cancer cell lines. The acute systemic toxicity profile of the nanogel (NG1) was studied in immunocompromised mice, indicating no signs of toxicity. Additionally, growth inhibition of subcutaneous FaDu xenograft tumors was observed with nanoIAZA, demonstrating that this nanoformulation offers a significant improvement in tumor regression and overall survival compared to the control.
Collapse
Affiliation(s)
- Diana Diaz-Dussan
- Department of Chemical & Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| | - Yi-Yang Peng
- Department of Chemical & Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| | - Faisal Bin Rashed
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, T6G 1Z2, Alberta, Canada
| | - Dawn Macdonald
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, T6G 1Z2, Alberta, Canada
| | - Michael Weinfeld
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, T6G 1Z2, Alberta, Canada
| | - Piyush Kumar
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, T6G 1Z2, Alberta, Canada
| | - Ravin Narain
- Department of Chemical & Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| |
Collapse
|
4
|
Fernández-Tabanera E, Melero-Fernández de Mera RM, Alonso J. CD44 In Sarcomas: A Comprehensive Review and Future Perspectives. Front Oncol 2022; 12:909450. [PMID: 35785191 PMCID: PMC9247467 DOI: 10.3389/fonc.2022.909450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 12/16/2022] Open
Abstract
It is widely accepted that the tumor microenvironment, particularly the extracellular matrix, plays an essential role in the development of tumors through the interaction with specific protein-membrane receptors. One of the most relevant proteins in this context is the transmembrane protein CD44. The role of CD44 in tumor progression, invasion, and metastasis has been well established in many cancers, although a comprehensive review concerning its role in sarcomas has not been published. CD44 is overexpressed in most sarcomas and several in vitro and in vivo experiments have shown a direct effect on tumor progression, dissemination, and drug resistance. Moreover, CD44 has been revealed as a useful marker for prognostic and diagnostic (CD44v6 isoform) in osteosarcoma. Besides, some innovative treatments such as HA-functionalized liposomes therapy have become an excellent CD44-mediated intracellular delivery system for osteosarcoma. Unfortunately, the reduced number of studies deciphering the prognostic/diagnostic value of CD44 in other sarcoma subgroups, neither than osteosarcoma, in addition to the low number of patients involved in those studies, have produced inconclusive results. In this review, we have gone through the information available on the role of CD44 in the development, maintenance, and progression of sarcomas, analyzing their implications at the prognostic, therapeutic, and mechanistic levels. Moreover, we illustrate how research involving the specific role of CD44 in the different sarcoma subgroups could suppose a chance to advance towards a more innovative perspective for novel therapies and future clinical trials.
Collapse
Affiliation(s)
- Enrique Fernández-Tabanera
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
- Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Raquel M. Melero-Fernández de Mera
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
| | - Javier Alonso
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
- *Correspondence: Javier Alonso,
| |
Collapse
|
5
|
Selective Viramidine-Loaded Aptamer-Nanoparticles Trigger Cell Cycle Arrest in Nucleolin-Expressed Hepatoma Cells Through Modulation of CDC25A/p53/PI3k Pathway. J CLUST SCI 2022. [DOI: 10.1007/s10876-022-02224-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
6
|
Salvati A, Poelstra K. Drug Targeting and Nanomedicine: Lessons Learned from Liver Targeting and Opportunities for Drug Innovation. Pharmaceutics 2022; 14:217. [PMID: 35057111 PMCID: PMC8777931 DOI: 10.3390/pharmaceutics14010217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/08/2023] Open
Abstract
Drug targeting and nanomedicine are different strategies for improving the delivery of drugs to their target. Several antibodies, immuno-drug conjugates and nanomedicines are already approved and used in clinics, demonstrating the potential of such approaches, including the recent examples of the DNA- and RNA-based vaccines against COVID-19 infections. Nevertheless, targeting remains a major challenge in drug delivery and different aspects of how these objects are processed at organism and cell level still remain unclear, hampering the further development of efficient targeted drugs. In this review, we compare properties and advantages of smaller targeted drug constructs on the one hand, and larger nanomedicines carrying higher drug payload on the other hand. With examples from ongoing research in our Department and experiences from drug delivery to liver fibrosis, we illustrate opportunities in drug targeting and nanomedicine and current challenges that the field needs to address in order to further improve their success.
Collapse
Affiliation(s)
- Anna Salvati
- Correspondence: (A.S.); (K.P.); Tel.: +31-503639831 (A.S.); +31-503633287 (K.P.)
| | - Klaas Poelstra
- Correspondence: (A.S.); (K.P.); Tel.: +31-503639831 (A.S.); +31-503633287 (K.P.)
| |
Collapse
|
7
|
Deng X, Wu Y, Xu H, Yan J, Liu H, Zhang B. Recent research progress in galactose-based fluorescent probes for detection of biomarkers of liver diseases. Chem Commun (Camb) 2022; 58:12518-12527. [DOI: 10.1039/d2cc04180d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This highlight illustrates the challenges and latest progress in galactose-based fluorescent probes for early diagnosis of liver diseases.
Collapse
Affiliation(s)
- Xiaojing Deng
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Yingxu Wu
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Hu Xu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 16044, China
| | - Jiawei Yan
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Huanying Liu
- School of Mechanical and Power Engineering, Dalian Ocean University, Dalian 116023, China
| | - Boyu Zhang
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
8
|
Singh D, Singh M. Hepatocellular-Targeted mRNA Delivery Using Functionalized Selenium Nanoparticles In Vitro. Pharmaceutics 2021; 13:298. [PMID: 33668320 PMCID: PMC8025905 DOI: 10.3390/pharmaceutics13030298] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023] Open
Abstract
Selenium's (Se) chemopreventative and therapeutic properties have attracted attention in nanomedicine. Se nanoparticles (SeNPs) retain these properties of Se while possessing lower toxicity and higher bioavailability, potentiating their use in gene delivery. This study aimed to formulate SeNPs for efficient binding and targeted delivery of FLuc-mRNA to hepatocellular carcinoma cells (HepG2) in vitro. The colorectal adenocarcinoma (Caco-2) and normal human embryonic kidney (HEK293) cells that do not have the asialoorosomucoid receptor (ASGPR) were utilized for comparison. SeNPs were functionalized with chitosan (CS), polyethylene glycol (PEG), and lactobionic acid (LA) for ASGPR targeting on HepG2 cells. Nanoparticles (NPs) and their mRNA-nanocomplexes were characterized by Fourier transform infra-red (FTIR) and UV-vis spectroscopy, transmission electron microscopy (TEM), and nanoparticle tracking analysis (NTA). Gel and fluorescence-based assays assessed the NP's ability to bind and protect FLuc-mRNA. Cytotoxicity was determined using the -(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, while transgene expression was evaluated using the luciferase reporter gene assay. All NPs appeared spherical with sizes ranging 57.2-130.0 nm and zeta potentials 14.9-31.4 mV. NPs bound, compacted, and protected the mRNA from nuclease digestion and showed negligible cytotoxicity in vitro. Targeted gene expression was highest in the HepG2 cells using the LA targeted NPs. These NPs portend to be efficient nanocarriers of nucleic acids and warrant further investigation.
Collapse
Affiliation(s)
| | - Moganavelli Singh
- Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, University of KwaZulu-Natal, Private Bag, Durban X54001, South Africa;
| |
Collapse
|
9
|
Diaz-Dussan D, Peng YY, Kumar P, Narain R. Oncogenic Epidermal Growth Factor Receptor Silencing in Cervical Carcinoma Mediated by Dynamic Sugar-Benzoxaborole Polyplexes. ACS Macro Lett 2020; 9:1464-1470. [PMID: 35653664 DOI: 10.1021/acsmacrolett.0c00599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although, various types of pharmaceuticals have been developed for cervical carcinomas, treatment with these drugs often results in a number of undesirable side effects, toxicity and multidrug resistance. Here, we aimed at modifying the genetic profiling of cancer cells by silencing the expression of the epidermal growth factor receptor (EGFR) gene. We have synthesized two kinds of RAFT-made, biocompatible, and cationic polymers for the encapsulation of silencing RNA (siRNA). This vector has a dual capability: it contains a cationic segment to complex with the siRNA and an omega-end modified with an oxaborole group via thiol-ene click chemistry that responds to the acidic tumor microenvironment. This structural innovation enables this macromolecule to interact with multiple polyplexes and release the siRNA in a mild acidic environment. A strategy that has shown enhanced gene silencing without elevating the cytotoxicity of the system, as determined by Western blot analysis. The success of this approach has afforded further interest in utilizing boron-carbohydrate interaction in the development of nonviral vectors for gene therapy.
Collapse
Affiliation(s)
- Diana Diaz-Dussan
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| | - Yi-Yang Peng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| | - Piyush Kumar
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, T6G 1Z2, Alberta, Canada
| | - Ravin Narain
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| |
Collapse
|
10
|
Siciliano G, Corricelli M, Iacobazzi RM, Canepa F, Comegna D, Fanizza E, Del Gatto A, Saviano M, Laquintana V, Comparelli R, Mascolo G, Murgolo S, Striccoli M, Agostiano A, Denora N, Zaccaro L, Curri ML, Depalo N. Gold-Speckled SPION@SiO 2 Nanoparticles Decorated with Thiocarbohydrates for ASGPR1 Targeting: Towards HCC Dual Mode Imaging Potential Applications. Chemistry 2020; 26:11048-11059. [PMID: 32628283 DOI: 10.1002/chem.202002142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Efforts are made to perform an early and accurate detection of hepatocellular carcinoma (HCC) by simultaneous exploiting multiple clinically non-invasive imaging modalities. Original nanostructures derived from the combination of different inorganic domains can be used as efficient contrast agents in multimodal imaging. Superparamagnetic iron oxide nanoparticles (SPIONs) and Au nanoparticles (NPs) possess well-established contrasting features in magnetic resonance imaging (MRI) and X-ray computed tomography (CT), respectively. HCC can be targeted by using specific carbohydrates able to recognize asialoglycoprotein receptor 1 (ASGPR1) overexpressed in hepatocytes. Here, two different thiocarbohydrate ligands were purposely designed and alternatively conjugated to the surface of Au-speckled silica-coated SPIONs NPs, to achieve two original nanostructures that could be potentially used for dual mode targeted imaging of HCC. The results indicated that the two thiocarbohydrate decorated nanostructures possess convenient plasmonic/superparamagnetic properties, well-controlled size and morphology and good selectivity for targeting ASGPR1 receptor.
Collapse
Affiliation(s)
- Giulia Siciliano
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy.,Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy.,Present address: Dipartimento di Matematica e Fisica "Ennio De Giorgi", Università del Salento, 73100, Lecce, Italy
| | - Michela Corricelli
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| | - Rosa Maria Iacobazzi
- Istituto Tumori Giovanni Paolo II, IRCCS, Viale Orazio Flacco 65, 70124, Bari, Italy
| | - Fabio Canepa
- Dipartimento di Chimica e Chimica Industriale-SPIN-CNR Unità di Genova, Università degli Studi di Genova, via Dodecaneso 31, 16146, Genova, Italy
| | - Daniela Comegna
- Istituto di Biostrutture e Bioimmagini IBB, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Elisabetta Fanizza
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy.,Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| | - Annarita Del Gatto
- Istituto di Biostrutture e Bioimmagini IBB, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Michele Saviano
- Istituto di Cristallografia IC, CNR, Via Giovanni Amendola, 122/O, 70126, Bari, Italy
| | - Valentino Laquintana
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| | - Roberto Comparelli
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| | - Giuseppe Mascolo
- Istituto di Ricerca Sulle Acque IRSA, CNR, Area della Ricerca Roma 1, Via Salaria Km 29,300 C.P. 10, 00015 Monterotondo Stazione, Roma, Italy
| | - Sapia Murgolo
- Istituto di Ricerca Sulle Acque IRSA, CNR, Viale Francesco de Blasio 5, 70132, Bari, Italy
| | - Marinella Striccoli
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| | - Angela Agostiano
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy.,Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| | - Nunzio Denora
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy.,Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| | - Laura Zaccaro
- Istituto di Biostrutture e Bioimmagini IBB, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - M Lucia Curri
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy.,Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| | - Nicoletta Depalo
- Istituto per i Processi Chimico Fisici IPCF S.S: Bari, CNR, Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, Via Orabona 4, 70124, Bari, Italy
| |
Collapse
|
11
|
Shetab Boushehri MA, Dietrich D, Lamprecht A. Nanotechnology as a Platform for the Development of Injectable Parenteral Formulations: A Comprehensive Review of the Know-Hows and State of the Art. Pharmaceutics 2020; 12:pharmaceutics12060510. [PMID: 32503171 PMCID: PMC7356945 DOI: 10.3390/pharmaceutics12060510] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Within recent decades, the development of nanotechnology has made a significant contribution to the progress of various fields of study, including the domains of medical and pharmaceutical sciences. A substantially transformed arena within the context of the latter is the development and production of various injectable parenteral formulations. Indeed, recent decades have witnessed a rapid growth of the marketed and pipeline nanotechnology-based injectable products, which is a testimony to the remarkability of the aforementioned contribution. Adjunct to the ability of nanomaterials to deliver the incorporated payloads to many different targets of interest, nanotechnology has substantially assisted to the development of many further facets of the art. Such contributions include the enhancement of the drug solubility, development of long-acting locally and systemically injectable formulations, tuning the onset of the drug’s release through the endowment of sensitivity to various internal or external stimuli, as well as adjuvancy and immune activation, which is a desirable component for injectable vaccines and immunotherapeutic formulations. The current work seeks to provide a comprehensive review of all the abovementioned contributions, along with the most recent advances made within each domain. Furthermore, recent developments within the domains of passive and active targeting will be briefly debated.
Collapse
Affiliation(s)
- Maryam A. Shetab Boushehri
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-736428; Fax: +49-228-735268
| | - Dirk Dietrich
- Department of Neurosurgery, University Clinic of Bonn, 53105 Bonn, Germany;
| | - Alf Lamprecht
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- PEPITE EA4267, Institute of Pharmacy, University Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
12
|
Liu Y, Tan M, Zhang Y, Huang W, Min L, Peng S, Yuan K, Qiu L, Min W. Targeted Gene Silencing BRAF Synergized Photothermal Effect Inhibits Hepatoma Cell Growth Using New GAL-GNR-siBRAF Nanosystem. NANOSCALE RESEARCH LETTERS 2020; 15:116. [PMID: 32449085 PMCID: PMC7246281 DOI: 10.1186/s11671-020-03340-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/01/2020] [Indexed: 05/04/2023]
Abstract
Liver cancer is one of the most common malignancies worldwide. The RAF kinase inhibitors are effective in the treatment of hepatocellular carcinoma (HCC); therefore, inhibition of the BRAF/MEK/ERK pathway has become a new therapeutic strategy for novel HCC therapy. However, targeted specific delivery systems for tumors are still significant obstacle to clinical applications. Galactose (GAL) can target the asialoglycoprotein receptor (ASGPR) that is highly expressed on liver cancer cells. In this study, we designed a novel multifunctional nanomaterial GAL-GNR-siBRAF which consists of three parts, GAL as the liver cancer-targeting moiety, golden nanorods (GNR) offering photothermal capability under near infrared light, and siRNA specifically silencing BRAF (siBRAF). The nanocarrier GAL-GNR-siBRAF showed high siRNA loading capacity and inhibited the degradation of siRNA in serum. Compared with naked gold nanorods, GAL-GNR-siBRAF possessed lower biotoxicity and higher efficacy of gene silencing. Treatment with GAL-GNR-siBRAF significantly downregulated the expression of BRAF and impaired proliferation, migration, and invasion of liver cancer cells. Moreover, combinatorial photothermal effects and BRAF knockdown by GAL-GNR-siBRAF effectively given rise to tumor cell death. Therefore, our study developed a new type of targeted multi-functional nanomaterial GAL-GNR-siBRAF for the treatment of liver cancer, which provides ideas for the development of new clinical treatment methods.
Collapse
Affiliation(s)
- Yanling Liu
- Institute of Immunotherapy, Nanchang University, Nanchang, 330006 Jiangxi China
- Jiangxi University of Technology, Nanchang, Jiangxi 330098 China
- Jiangxi Institute of Medical Sciences Nanchang, Nanchang, 330006 Jiangxi China
| | - Manman Tan
- Institute of Immunotherapy, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Yujuan Zhang
- Institute of Immunotherapy, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Wei Huang
- Institute of Immunotherapy, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Liangliang Min
- Institute of Immunotherapy, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Shanshan Peng
- Jiangxi Institute of Medical Sciences Nanchang, Nanchang, 330006 Jiangxi China
| | - Keng Yuan
- Jiangxi Institute of Medical Sciences Nanchang, Nanchang, 330006 Jiangxi China
| | - Li Qiu
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, China
| | - Weiping Min
- Institute of Immunotherapy, Nanchang University, Nanchang, 330006 Jiangxi China
- Jiangxi University of Technology, Nanchang, Jiangxi 330098 China
- Jiangxi Institute of Medical Sciences Nanchang, Nanchang, 330006 Jiangxi China
- Department of Surgery, Pathology and Oncology, University of Western Ontario, London, N6A 5A5 Canada
| |
Collapse
|
13
|
Reshitko GS, Yamansarov EY, Evteev SA, Lopatukhina EV, Shkil' DO, Saltykova IV, Lopukhov AV, Kovalev SV, Lobov AN, Kislyakov IV, Burenina OY, Klyachko NL, Garanina AS, Dontsova OA, Ivanenkov YA, Erofeev AS, Gorelkin PV, Beloglazkina EK, Majouga AG. Synthesis and Evaluation of New Trivalent Ligands for Hepatocyte Targeting via the Asialoglycoprotein Receptor. Bioconjug Chem 2020; 31:1313-1319. [PMID: 32379426 DOI: 10.1021/acs.bioconjchem.0c00202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the asialoglycoprotein receptor (also known as the "Ashwell-Morell receptor" or ASGPR) was discovered as the first cellular mammalian lectin, numerous drug delivery systems have been developed and several gene delivery systems associated with multivalent ligands for liver disease targeting are undergoing clinical trials. The success of these systems has facilitated the further study of new ligands with comparable or higher affinity and less synthetic complexity. Herein, we designed two novel trivalent ligands based on the esterification of tris(hydroxymethyl) aminomethane (TRIS) followed by the azide-alkyne Huisgen cycloaddition with azido N-acetyl-d-galactosamine. The presented triazolyl glycoconjugates exhibited good binding to ASGPR, which was predicted using in silico molecular docking and assessed by a surface plasmon resonance (SPR) technique. Moreover, we demonstrated the low level of in vitro cytotoxicity, as well as the optimal spatial geometry and the required amphiphilic balance, for new, easily accessible ligands. The conjugate of a new ligand with Cy5 dye exhibited selective penetration into HepG2 cells in contrast to the ASGPR-negative PC3 cell line.
Collapse
Affiliation(s)
- Galina S Reshitko
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Emil Yu Yamansarov
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Sergei A Evteev
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Elena V Lopatukhina
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Dmitry O Shkil'
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Irina V Saltykova
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Anton V Lopukhov
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Sergey V Kovalev
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Alexander N Lobov
- Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, 450054, Russian Federation
| | - Ivan V Kislyakov
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Olga Yu Burenina
- Skolkovo Institute of Science and Technology, Skolkovo, 143026, Russian Federation
| | - Natalia L Klyachko
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,Skolkovo Institute of Science and Technology, Skolkovo, 143026, Russian Federation
| | - Anastasiia S Garanina
- National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Olga A Dontsova
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,Skolkovo Institute of Science and Technology, Skolkovo, 143026, Russian Federation
| | - Yan A Ivanenkov
- Moscow Institute of Physics and Technology (State University), Dolgoprudny City, Moscow Region 141700, Russian Federation.,Institute of Biochemistry and Genetics, Russian Academy of Science (IBG RAS) of the Ufa Federal Research Centre, Ufa, 450054, Russian Federation
| | - Alexander S Erofeev
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Peter V Gorelkin
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Elena K Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Alexander G Majouga
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow, 119049, Russian Federation.,Dmitry Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation
| |
Collapse
|
14
|
Nanoparticles Based on Novel Carbohydrate-Functionalized Polymers. Molecules 2020; 25:molecules25071744. [PMID: 32290160 PMCID: PMC7180923 DOI: 10.3390/molecules25071744] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/05/2022] Open
Abstract
Polymeric nanoparticles can be used for drug delivery systems in healthcare. For this purpose poly(lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) offer an excellent polymeric matrix. In this work, PLGA and PEG polymers were functionalized with coumarin and carbohydrate moieties such as thymidine, glucose, galactose, and mannose that have high biological specificities. Using a single oil in water emulsion methodology, functionalized PLGA nanoparticles were prepared having a smooth surface and sizes ranging between 114–289 nm, a low polydispersity index and a zeta potential from −28.2 to −56.0 mV. However, for the corresponding PEG derivatives the polymers obtained were produced in the form of films due to the small size of the hydrophobic core.
Collapse
|
15
|
Martínez-Negro M, Sánchez-Arribas N, Guerrero-Martínez A, Moyá ML, Tros de Ilarduya C, Mendicuti F, Aicart E, Junquera E. A Non-Viral Plasmid DNA Delivery System Consisting on a Lysine-Derived Cationic Lipid Mixed with a Fusogenic Lipid. Pharmaceutics 2019; 11:E632. [PMID: 31783620 PMCID: PMC6956073 DOI: 10.3390/pharmaceutics11120632] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022] Open
Abstract
The insertion of biocompatible amino acid moieties in non-viral gene nanocarriers is an attractive approach that has been recently gaining interest. In this work, a cationic lipid, consisting of a lysine-derived moiety linked to a C12 chain (LYCl) was combined with a common fusogenic helper lipid (DOPE) and evaluated as a potential vehicle to transfect two plasmid DNAs (encoding green fluorescent protein GFP and luciferase) into COS-7 cells. A multidisciplinary approach has been followed: (i) biophysical characterization based on zeta potential, gel electrophoresis, small-angle X-ray scattering (SAXS), and cryo-transmission electronic microscopy (cryo-TEM); (ii) biological studies by fluorescence assisted cell sorting (FACS), luminometry, and cytotoxicity experiments; and (iii) a computational study of the formation of lipid bilayers and their subsequent stabilization with DNA. The results indicate that LYCl/DOPE nanocarriers are capable of compacting the pDNAs and protecting them efficiently against DNase I degradation, by forming Lα lyotropic liquid crystal phases, with an average size of ~200 nm and low polydispersity that facilitate the cellular uptake process. The computational results confirmed that the LYCl/DOPE lipid bilayers are stable and also capable of stabilizing DNA fragments via lipoplex formation, with dimensions consistent with experimental values. The optimum formulations (found at 20% of LYCl content) were able to complete the transfection process efficiently and with high cell viabilities, even improving the outcomes of the positive control Lipo2000*.
Collapse
Affiliation(s)
- María Martínez-Negro
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.M.-N.); (N.S.-A.); (A.G.-M.); (E.A.)
| | - Natalia Sánchez-Arribas
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.M.-N.); (N.S.-A.); (A.G.-M.); (E.A.)
| | - Andrés Guerrero-Martínez
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.M.-N.); (N.S.-A.); (A.G.-M.); (E.A.)
| | - María Luisa Moyá
- Grupo de Química Coloidal y Catálisis Micelar, Departamento de Química Física, Facultad de Química, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - Conchita Tros de Ilarduya
- Departamento de Tecnología y Química Farmacéuticas, Facultad de Farmacia y Nutrición, Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31080 Pamplona, Spain;
| | - Francisco Mendicuti
- Departmento de Química Analítica, Química Física e Ingeniería Química and Instituto de Investigación Quimica Andrés M. del Rio, Universidad de Alcalá, 28871 Alcalá de Henares, Spain;
| | - Emilio Aicart
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.M.-N.); (N.S.-A.); (A.G.-M.); (E.A.)
| | - Elena Junquera
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.M.-N.); (N.S.-A.); (A.G.-M.); (E.A.)
| |
Collapse
|
16
|
Gauthier L, Varache M, Couffin AC, Lebrun C, Delangle P, Gateau C, Texier I. Quantification of Surface GalNAc Ligands Decorating Nanostructured Lipid Carriers by UPLC-ELSD. Int J Mol Sci 2019; 20:ijms20225669. [PMID: 31726778 PMCID: PMC6888163 DOI: 10.3390/ijms20225669] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/07/2019] [Accepted: 11/09/2019] [Indexed: 12/16/2022] Open
Abstract
Nanoparticles have been extensively studied for drug delivery and targeting to specific organs. The functionalization of the nanoparticle surface by site-specific ligands (antibodies, peptides, saccharides) can ensure efficient recognition and binding with relevant biological targets. One of the main challenges in the development of these decorated nanocarriers is the accurate quantification of the amount of ligands on the nanoparticle surface. In this study, nanostructured lipid carriers (NLC) were functionalized with N-acetyl-D-galactosamine (GalNAc) units, known to target the asialoglycoprotein receptor (ASGPR). Different molar percentages of GalNAc-functionalized surfactant (0%, 2%, 5%, and 14%) were used in the formulation. Based on ultra-high-performance liquid chromatography separation and evaporative light-scattering detection (UPLC-ELSD), an analytical method was developed to specifically quantify the amount of GalNAc units present at the NLC surface. This method allowed the accurate quantification of GalNAc surfactant and therefore gave some insights into the structural parameters of these multivalent ligand systems. Our data show that the GalNAc decorated NLC possess large numbers of ligands at their surface and suitable distances between them for efficient multivalent interaction with the ASGPR, and therefore promising liver-targeting efficiency.
Collapse
Affiliation(s)
- Laura Gauthier
- Université Grenoble Alpes, CEA, LETI-DTBS, F-38000 Grenoble, France; (L.G.); (M.V.); (A.-C.C.)
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, F-38000 Grenoble, France; (C.L.); (P.D.)
| | - Mathieu Varache
- Université Grenoble Alpes, CEA, LETI-DTBS, F-38000 Grenoble, France; (L.G.); (M.V.); (A.-C.C.)
| | - Anne-Claude Couffin
- Université Grenoble Alpes, CEA, LETI-DTBS, F-38000 Grenoble, France; (L.G.); (M.V.); (A.-C.C.)
| | - Colette Lebrun
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, F-38000 Grenoble, France; (C.L.); (P.D.)
| | - Pascale Delangle
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, F-38000 Grenoble, France; (C.L.); (P.D.)
| | - Christelle Gateau
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, F-38000 Grenoble, France; (C.L.); (P.D.)
- Correspondence: (C.G.); (I.T.); Tel.: +33-438-786-041 (C.G.); +33-438-784-670 (I.T.)
| | - Isabelle Texier
- Université Grenoble Alpes, CEA, LETI-DTBS, F-38000 Grenoble, France; (L.G.); (M.V.); (A.-C.C.)
- Correspondence: (C.G.); (I.T.); Tel.: +33-438-786-041 (C.G.); +33-438-784-670 (I.T.)
| |
Collapse
|
17
|
Hofmann S, Bellmann-Sickert K, Beck-Sickinger AG. Chemical modification of neuropeptide Y for human Y1 receptor targeting in health and disease. Biol Chem 2019; 400:299-311. [PMID: 30653463 DOI: 10.1515/hsz-2018-0364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022]
Abstract
As a very abundant neuropeptide in the brain and widely distributed peptide hormone in the periphery, neuropeptide Y (NPY) appears to be a multisignaling key peptide. Together with peptide YY, pancreatic polypeptide and the four human G protein-coupled receptor subtypes hY1R, hY2R, hY4R and hY5R it forms the NPY/hYR multiligand/multireceptor system, which is involved in essential physiological processes as well as in human diseases. In particular, NPY-induced hY1R signaling plays a central role in the regulation of food intake and stress response as well as in obesity, mood disorders and cancer. Thus, several hY1R-preferring NPY analogs have been developed as versatile tools to unravel the complex NPY/hY1R signaling in health and disease. Further, these peptides provide basic lead structures for the development of innovative drugs. Here, the current research is summarized focusing on the development of differently sized hY1R-preferring NPY analogs as well as their advances with respect to hY1R profiling, potential therapeutic applications and targeted cancer imaging and therapy. Finally, major limitations and innovative strategies for next generation hY1R-preferring NPY analogs are addressed.
Collapse
Affiliation(s)
- Sven Hofmann
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| | - Kathrin Bellmann-Sickert
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| |
Collapse
|
18
|
Chen F, Huang G. Application of glycosylation in targeted drug delivery. Eur J Med Chem 2019; 182:111612. [DOI: 10.1016/j.ejmech.2019.111612] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/26/2019] [Accepted: 08/09/2019] [Indexed: 01/10/2023]
|
19
|
Barclay TG, Day CM, Petrovsky N, Garg S. Review of polysaccharide particle-based functional drug delivery. Carbohydr Polym 2019; 221:94-112. [PMID: 31227171 PMCID: PMC6626612 DOI: 10.1016/j.carbpol.2019.05.067] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 01/06/2023]
Abstract
This review investigates the significant role polysaccharide particles play in functional drug delivery. The importance of these systems is due to the wide variety of polysaccharides and their natural source meaning that they can provide biocompatible and biodegradable systems with a range of both biological and chemical functionality valuable for drug delivery. This functionality includes protection and presentation of working therapeutics through avoidance of the reticuloendothelial system, stabilization of biomacromolecules and increasing the bioavailability of incorporated small molecule drugs. Transport of the therapeutic is also key to the utility of polysaccharide particles, moving drugs from the site of administration through mucosal binding and transport and using chemistry, size and receptor mediated drug targeting to specific tissues. This review also scrutinizes the methods of synthesizing and constructing functional polysaccharide particle drug delivery systems that maintain and extend the functionality of the natural polysaccharides.
Collapse
Affiliation(s)
- Thomas G Barclay
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia.
| | - Candace Minhthu Day
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia.
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 1 Flinders Drive, Bedford Park, SA 5042, Australia; Department of Endocrinology, Flinders Medical Centre/Flinders University, Bedford Park, SA 5042, Australia.
| | - Sanjay Garg
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
20
|
Abd-Rabou AA, Bharali DJ, Mousa SA. Viramidine-Loaded Galactosylated Nanoparticles Induce Hepatic Cancer Cell Apoptosis and Inhibit Angiogenesis. Appl Biochem Biotechnol 2019; 190:305-324. [PMID: 31346920 DOI: 10.1007/s12010-019-03090-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/05/2019] [Indexed: 01/19/2023]
Abstract
Current estimates indicate that hepatocarcinoma is the leading cause of death globally. There is interest in utilizing nanomedicine for cancer therapy to overcome side effects of chemo-interventions. Ribavirin, an antiviral nucleoside inhibitor, accumulates inside red blood cells, causing anemia. Its analog, viramidine, can concentrate within hepatocytes and spare red blood cells, thus limiting anemia. Hepatocarcinoma cells have a large number of asialoglycoprotein receptors on their membranes that can bind galactosyl-terminating solid lipid nanoparticles (Gal-SLN) and internalize them. Here, viramidine, 5-fluorouracil, and paclitaxel-loaded Gal-SLN were characterized inside cells. Cytotoxicities of free-drug, nano-void, and drug-loaded Gal-SLN were evaluated using HepG2 cells; over 3 days, cell viability was measured. To test the mechanistic pathway, we investigated in vitro apoptosis using flow cytometry and in ovo angiogenesis using the CAM assay. Results showed that 1 and 2 μM of the viramidine-encapsulated Gal-SLN had the highest cytotoxic effect, achieving 80% cell death with a steady increase over 3 days, with induction of apoptosis and reduction of necrosis and angiogenesis, compared to free-drugs. Gal-SLN application on breast cancer MCF-7 cells confirmed its specificity against liver cancer HepG2 cells. We conclude that viramidine-encapsulated Gal-SLN has anticancer and anti-angiogenic activities against hepatocarcinoma.
Collapse
Affiliation(s)
- Ahmed A Abd-Rabou
- Hormones Department, Medical Research Division, National Research Centre, Giza, 12622, Egypt.,Stem Cell Laboratory, Center of Excellence for Advanced Science, National Research Centre, Giza, 12622, Egypt
| | - Dhruba J Bharali
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, 12144, USA
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, 12144, USA.
| |
Collapse
|
21
|
Yousef S, Alsaab HO, Sau S, Iyer AK. Development of asialoglycoprotein receptor directed nanoparticles for selective delivery of curcumin derivative to hepatocellular carcinoma. Heliyon 2018; 4:e01071. [PMID: 30603704 PMCID: PMC6305692 DOI: 10.1016/j.heliyon.2018.e01071] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/11/2018] [Accepted: 12/17/2018] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular cellular carcinoma (HCC) is one of the most challenging liver cancer subtypes. Due to lack of cell surface biomarkers and highly metastatic nature, early detection and targeted therapy of HCC is an unmet need. Galactosamine (Gal) is among the few selective ligands used for targeting HCCs due to its high binding affinity to asialoglycoprotein receptors (ASGPRs) overexpressed in HCC. In the present work, we engineered nanoscale G4 polyamidoamine (PAMAM) dendrimers anchored to galactosamine and loaded with the potent anticancer curcumin derivative (CDF) as a platform for targeted drug delivery to HCC. In vivo targeting ability and bio-distribution of PAMAM-Gal were assessed via its labeling with the clinically used, highly contrast, near infrared (NIR) dye: S0456, with testing of the obtained conjugate in aggressive HCC xenograft model. Our results highlighted the targeted dendrimer PAMAM-Gal ability to achieve selective high cellular uptake via ASGPR mediated endocytosis and significantly enhance the delivery of CDF into the studied HCC cell lines. Cytotoxicity MTT assays in HCC cell lines, interestingly highlighted, the comparative high potency of CDF, where CDF was more potent as a chemotherapeutic anticancer small molecule than the currently in use Doxorubicin, Sorafenib and Cisplatin chemotherapeutic agents. In conclusion the proof-of-concept study using nanoscale PAMAM-Gal dendrimer has demonstrated its competency as an efficient delivery system for selective delivery of potent CDF for HCC anticancer therapy as well as HCC diagnosis via NIR imaging.
Collapse
Affiliation(s)
- Shaimaa Yousef
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Hashem O. Alsaab
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samaresh Sau
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Arun K. Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Molecular Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
22
|
Hu J, Wei P, Seeberger PH, Yin J. Mannose-Functionalized Nanoscaffolds for Targeted Delivery in Biomedical Applications. Chem Asian J 2018; 13:3448-3459. [PMID: 30251341 DOI: 10.1002/asia.201801088] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/18/2018] [Indexed: 12/27/2022]
Abstract
Targeted drug delivery by nanomaterials has been extensively investigated as an effective strategy to surmount obstacles in the conventional treatment of cancer and infectious diseases, such as systemic toxicity, low drug efficacy, and drug resistance. Mannose-binding C-type lectins, which primarily include mannose receptor (MR, CD206) and dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), are highly expressed on various cancer cells, endothelial cells, macrophages, and dendritic cells (DCs), which make them attractive targets for therapeutic effect. Mannosylated nanomaterials hold great potential in cancer and infection treatment on account of their direct therapeutic effect on targeted cells, modulation of the tumor microenvironment, and stimulation of immune response through antigen presentation. This review presents the recent advances in mannose-based targeted delivery nanoplatforms incorporated with different therapies in the biomedical field.
Collapse
Affiliation(s)
- Jing Hu
- Wuxi School of Medicine, Jiangnan University, Lihu Avenue1800, Wuxi, 214122, China
| | - Peng Wei
- Department Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education, School of Biotechnology, Jiangnan University, Lihu Avenue1800, Wuxi, 214122, China
| | - Peter H Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476, Potsdam, Germany
| | - Jian Yin
- Department Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education, School of Biotechnology, Jiangnan University, Lihu Avenue1800, Wuxi, 214122, China
| |
Collapse
|
23
|
Zheng Y, Ji X, Yu B, Ji K, Gallo D, Csizmadia E, Zhu M, Choudhury MR, De La Cruz LKC, Chittavong V, Pan Z, Yuan Z, Otterbein LE, Wang B. Enrichment-triggered prodrug activation demonstrated through mitochondria-targeted delivery of doxorubicin and carbon monoxide. Nat Chem 2018; 10:787-794. [PMID: 29760413 PMCID: PMC6235738 DOI: 10.1038/s41557-018-0055-2] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/29/2018] [Indexed: 02/08/2023]
Abstract
Controlled activation is a critical component in prodrug development. Here we report a concentration-sensitive platform approach for bioorthogonal prodrug activation by taking advantage of reaction kinetics. Using two 'click and release' systems, we demonstrate enrichment and prodrug activation specifically in mitochondria to demonstrate the principle of the approach. In both cases, the payload (doxorubicin or carbon monoxide) was released inside the mitochondrial matrix following the enrichment-initiated click reaction. Furthermore, mitochondria-targeted delivery yielded substantial augmentation of functional biological and therapeutic effects in vitro and in vivo when compared to controls, which did not result in enrichment. This method is thus a platform for targeted drug delivery that is amenable to conjugation with a variety of molecules and is not limited to cell-surface delivery. Taken together, these two 'click and release' pairs clearly demonstrate the concept of enrichment-triggered drug release and the critical feasibility of treating clinically relevant diseases such as acute liver injury and cancer.
Collapse
Affiliation(s)
- Yueqin Zheng
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Xingyue Ji
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Bingchen Yu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Kaili Ji
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - David Gallo
- Harvard Medical School, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Eva Csizmadia
- Harvard Medical School, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mengyuan Zhu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Manjusha Roy Choudhury
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Ladie Kimberly C De La Cruz
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Vayou Chittavong
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Zhixiang Pan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Zhengnan Yuan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Leo E Otterbein
- Harvard Medical School, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA.
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
24
|
Potential therapeutic application of dendrimer/cyclodextrin conjugates with targeting ligands as advanced carriers for gene and oligonucleotide drugs. Ther Deliv 2017; 8:215-232. [PMID: 28222660 DOI: 10.4155/tde-2016-0064] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Despite the recent approval of some gene medicines and nucleic acid drugs, further improvement of delivery techniques for these drugs is strongly required. Several delivery technologies for these drugs have been developed, in other words, viral and two types of nonviral (lipofection and polyfection) vectors. Among the polyfection system, the potential use of various cyclodextrin (CyD) derivatives and CyD-appended polymers as carriers for gene and nucleic acid drugs has been demonstrated. The polyamidoamine dendrimer (G3) conjugates with α-CyD (α-CDE (G3)) have been reported to possess noteworthy properties as DNA and nucleic acid drugs carriers. This review will focus on the attempts to develop such cell-specific drug carriers by preparing polyethylene glycol, galactose, lactose, mannose, fucose and folic acid-appended α-CDEs as tissue and cell-selective carriers of gene and nucleic acid drugs.
Collapse
|
25
|
Gao YY, Chen H, Zhou YY, Wang LT, Hou Y, Xia XH, Ding Y. Intraorgan Targeting of Gold Conjugates for Precise Liver Cancer Treatment. ACS APPLIED MATERIALS & INTERFACES 2017; 9:31458-31468. [PMID: 28838233 DOI: 10.1021/acsami.7b08969] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Intraorgan targeting of chemical drugs at tumor tissues is essential in the treatment of solid tumors that express the same target receptor as normal tissues. Here, asialoglycoprotein receptor (ASGP-R)-targeting paclitaxel-conjugated gold nanoparticles (Gal/PTX-GNPs) are fabricated as a demonstration to realize the precise treatment of liver cancer. The enhanced biological specificity and therapeutic performance of drugs loaded on nanoparticles not only rely on the ligands on carriers for receptor recognition but are also determined by the performance of gold conjugates with designed structure. The tumor cell selectivity of the designed conjugates in liver tumor (HepG2) cells is close to six times of that incubated with control conjugates without galactose modification in liver normal (L02) cells. The drug level in tumor versus liver of Gal/PTX-GNPs is 121.0% at 8 h post injection, a 15.7-fold increase in the tumor specificity compared to that of GNPs conjugated with PTX only. This intraorgan-targeting strategy results in a considerable improvement of performance in treating both Heps heterotopic and orthotopic xenograft tumor models, which is expected to be used for the enhanced antitumor efficacy and reduced hepatotoxicity in liver cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | - Yanglong Hou
- Department of Materials Science and Engineering, College of Engineering and Beijing Key Laboratory for Magnetoelectric Materials and Devices, Peking University , Beijing 100871, China
| | | | | |
Collapse
|
26
|
Tsend-Ayush A, Zhu X, Ding Y, Yao J, Yin L, Zhou J, Yao J. Lactobionic acid-conjugated TPGS nanoparticles for enhancing therapeutic efficacy of etoposide against hepatocellular carcinoma. NANOTECHNOLOGY 2017; 28:195602. [PMID: 28291743 DOI: 10.1088/1361-6528/aa66ba] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Many effective anti-cancer drugs have limited use in hepatocellular carcinoma (HCC) therapy due to the drug resistance mechanisms in liver cells. In recent years, tumor-targeted drug delivery and the inhibition of drug-resistance-related mechanisms has become an integrated strategy for effectively combating chemo-resistant cancer. Herein, lactobionic acid-conjugated d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS-LA conjugate) has been developed as a potential asialoglycoprotein receptor (ASGPR)-targeted nanocarrier and an efficient inhibitor of P-glycoprotein (P-gp) to enhance etoposide (ETO) efficacy against HCC. The main properties of ETO-loaded TPGS-LA nanoparticles (NPs) were tested through in vitro and in vivo studies after being prepared using the nanoprecipitation method and characterized by dynamic light scattering (DLS). According to the results, smaller (∼141.43 nm), positively charged ETO-loaded TPGS-LA NPs were more suitable for providing efficient delivery to hepatoma cells by avoiding the clearance mechanisms. It was found that ETO-loaded TPGS-LA NPs were noticeably able to enhance the cytotoxicity of ETO in HepG2 cells. Besides this, markedly higher internalization by the ASGPR-overexpressed HepG2 cells and efficient accumulation at the tumor site in vivo were revealed in the TPGS-LA NP group. More importantly, animal studies confirmed that ETO-loaded TPGS-LA NPs achieved the highest therapeutic efficacy against HCC. Interestingly, ETO-loaded TPGS-LA NPs also exhibited a great inhibitory effect on P-gp compared to the ETO-loaded TPGS NPs. These results suggest that TPGS-LA NPs could be used as a potential ETO delivery system against HCC.
Collapse
Affiliation(s)
- Altansukh Tsend-Ayush
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
27
|
Wickens JM, Alsaab HO, Kesharwani P, Bhise K, Amin MCIM, Tekade RK, Gupta U, Iyer AK. Recent advances in hyaluronic acid-decorated nanocarriers for targeted cancer therapy. Drug Discov Today 2017; 22:665-680. [PMID: 28017836 PMCID: PMC5413407 DOI: 10.1016/j.drudis.2016.12.009] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/28/2016] [Accepted: 12/15/2016] [Indexed: 12/16/2022]
Abstract
The cluster-determinant 44 (CD44) receptor has a high affinity for hyaluronic acid (HA) binding and is a desirable receptor for active targeting based on its overexpression in cancer cells compared with normal body cells. The nanocarrier affinity can be increased by conjugating drug-loaded carriers with HA, allowing enhanced cancer cell uptake via the HA-CD44 receptor-mediated endocytosis pathway. In this review, we discuss recent advances in HA-based nanocarriers and micelles for cancer therapy. In vitro and in vivo experiments have repeatedly indicated HA-based nanocarriers to be a target-specific drug and gene delivery platform with great promise for future applications in clinical cancer therapy.
Collapse
Affiliation(s)
- Jennifer M Wickens
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; Department of Chemistry, Lake Superior State University, 680 W. Easterday Avenue, Sault Ste. Marie, MI 49783, USA
| | - Hashem O Alsaab
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA
| | - Prashant Kesharwani
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; The International Medical University, School of Pharmacy, Department of Pharmaceutical Technology, Jalan Jalil Perkasa 19, 57000 Kuala Lumpur, Malaysia
| | - Ketki Bhise
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education and Research (NIPER), Sarkhej - Gandhinagar Highway, Thaltej, Ahmedabad 380054, Gujarat, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
28
|
Insight into the role of dual-ligand modification in low molecular weight heparin based nanocarrier for targeted delivery of doxorubicin. Int J Pharm 2017; 523:427-438. [PMID: 28359815 DOI: 10.1016/j.ijpharm.2017.03.065] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/15/2017] [Accepted: 03/26/2017] [Indexed: 12/26/2022]
Abstract
Low molecular weight heparin nanoparticles (LMWH) modified by glycyrrhetinic acid (GA) (LMWH-GA) and further decorated by lactobionic acid (LA) (LA-LMWH-GA) were reported as novel hepatocellular carcinoma (HPC)-targeted carriers to overcome multidrug resistance (MDR) of doxorubicin (DOX). The drug-loaded nanoparticles had negative charge of around -25mV and average size range of 70-170nm. These nanoparticles performed sustained drug release in vitro and prolonged DOX residence time in blood circulation in vivo. Compared to free DOX, DOX-loaded nanoparticles demonstrated increased DOX accumulation in drug-resistance HepG2/ADR cells and enhanced in vitro therapeutic efficacy. However, DOX/LA-LMWH-GA with dual ligands didn't show higher cellular uptake and cytotoxicity than single GA modified DOX/LMWH-GA, although both GA-mediated and LA-mediated endocytosis were involved in their cell internalization. Uptake pathway inhibition study revealed the less efficacy of DOX/LA-LMWH-GA in cellular level could be attributed to the reduced effect of micropinocytosis and caveolae-mediated endocytosis in cellular uptake. Interestingly, the DOX-loaded nanoparticles developed from lower drug/carrier feeding ratio possessed higher performance in cell internalization and in vitro efficacy compared to those developed from higher drug/carrier feeding ratio, which could highlight the role of carrier in drug delivery process.
Collapse
|
29
|
Kotsuchibashi Y, Lee CM, Constantinescu I, Takeuchi LE, Vappala S, Kizhakkedathu JN, Narain R, Ebara M, Aoyagi T. A nanoparticle-preparation kit using ethylene glycol-based block copolymers with a common temperature-responsive block. Polym Chem 2017. [DOI: 10.1039/c7py01541k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A nanoparticle-preparation kit system using ethylene glycol-based block copolymers with a common temperature-responsive block was developed.
Collapse
Affiliation(s)
- Yohei Kotsuchibashi
- Department of Materials and Life Science
- Shizuoka Institute of Science and Technology
- Fukuroi
- Japan
- International Center for Materials Nanoarchitectonics (WPI-MANA)
| | - Chun Man Lee
- Medical Center for Translational Research
- Osaka University Hospital
- Suita
- Japan
| | - Iren Constantinescu
- Centre for Blood Research
- Department of Pathology and Laboratory Medicine
- University of British Columbia
- Vancouver
- Canada
| | - Lily E. Takeuchi
- Centre for Blood Research
- Department of Pathology and Laboratory Medicine
- University of British Columbia
- Vancouver
- Canada
| | - Sreeparna Vappala
- Centre for Blood Research
- Department of Pathology and Laboratory Medicine
- University of British Columbia
- Vancouver
- Canada
| | - Jayachandran N. Kizhakkedathu
- Centre for Blood Research
- Department of Pathology and Laboratory Medicine
- University of British Columbia
- Vancouver
- Canada
| | - Ravin Narain
- Department of Chemical and Materials Engineering
- University of Alberta
- Edmonton
- Canada
| | - Mitsuhiro Ebara
- International Center for Materials Nanoarchitectonics (WPI-MANA)
- National Institute for Materials Science (NIMS)
- Tsukuba
- Japan
- Materials and Science Engineering
| | - Takao Aoyagi
- Department of Materials and Applied Chemistry
- Nihon University
- Chiyoda-ku
- Japan
| |
Collapse
|
30
|
Hagimori M, Fuchigami Y, Kawakami S. Peptide-Based Cancer-Targeted DDS and Molecular Imaging. Chem Pharm Bull (Tokyo) 2017; 65:618-624. [DOI: 10.1248/cpb.c17-00098] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masayori Hagimori
- Department of Pharmaceutical Informatics, Nagasaki University Graduate School of Biomedical Sciences
| | - Yuki Fuchigami
- Department of Pharmaceutical Informatics, Nagasaki University Graduate School of Biomedical Sciences
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Nagasaki University Graduate School of Biomedical Sciences
| |
Collapse
|
31
|
Jefferis R. Recombinant Proteins and Monoclonal Antibodies. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2017; 175:281-318. [DOI: 10.1007/10_2017_32] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
32
|
Huang X, Leroux JC, Castagner B. Well-Defined Multivalent Ligands for Hepatocytes Targeting via Asialoglycoprotein Receptor. Bioconjug Chem 2016; 28:283-295. [DOI: 10.1021/acs.bioconjchem.6b00651] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Xiangang Huang
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Bastien Castagner
- Department
of Pharmacology and Therapeutics, McGill University, 3655 Prom. Sir-William-Osler, Montréal, Québec H3G 1Y6, Canada
| |
Collapse
|
33
|
He C, Wang S, Liu M, Zhao C, Xiang S, Zeng Y. Design, synthesis and in vitro evaluation of d-glucose-based cationic glycolipids for gene delivery. Org Biomol Chem 2016; 14:1611-22. [PMID: 26670704 DOI: 10.1039/c5ob02107c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A cationic lipid consists of a hydrophilic headgroup, backbone and hydrophobic tails which have an immense influence on the transfection efficiency of the lipid. In this paper, two novel series of cationic cyclic glycolipids with a quaternary ammonium headgroup and different-length hydrophobic tails (dodecyl, tetradecyl, hexadecyl) have been designed and synthesized for gene delivery. One contains lipids 1-3 with two hydrophobic alkyl chains linked to the glucose ring directly via an ether link. The other contains lipids 4-6 with two hydrophobic chains on the positively charged nitrogen atoms. All of the lipids were characterized for their ability to bind to DNA, size, ζ-potential, and toxicity. Atomic force microscopy showed that the lipids and DNA-lipid complexes were sphere-like forms. The lipids were used to transfer enhanced green fluorescent protein (EGFP-C3) to HEK293 cells without a helper lipid, the results indicated that lipids 4-6 have better transfection efficiency, in particular lipids 5-6 have similar or better efficiency, compared with the commercial transfection reagent lipofectamine 2000.
Collapse
Affiliation(s)
- Chengxi He
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, Hunan Normal University, Changsha, Hunan 410081, P. R. China.
| | - Shang Wang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P. R. China
| | - Meiyan Liu
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, Hunan Normal University, Changsha, Hunan 410081, P. R. China.
| | - Chunyan Zhao
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, Hunan Normal University, Changsha, Hunan 410081, P. R. China.
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P. R. China
| | - Youlin Zeng
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, Hunan Normal University, Changsha, Hunan 410081, P. R. China.
| |
Collapse
|
34
|
Leite JP, Mota R, Durão J, Neves SC, Barrias CC, Tamagnini P, Gales L. Cyanobacterium-Derived Extracellular Carbohydrate Polymer for the Controlled Delivery of Functional Proteins. Macromol Biosci 2016; 17. [PMID: 27594050 DOI: 10.1002/mabi.201600206] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/29/2016] [Indexed: 11/11/2022]
Abstract
The unicellular cyanobacterium Cyanothece sp. CCY 0110 is a highly efficient producer of extracellular polymeric substances (EPS), releasing up to 75% of the polymer to the culture medium. The carbohydrate polymer released to the medium (RPS) was previously isolated and characterized; it is composed of nine different monosaccharides including two uronic acids, and also containing peptides and sulfate groups. Here it is shown that the RPS spontaneously assembles with proteins at high concentrations leading to a phase transition. The proteins are released progressively and structurally intact near physiological conditions, primarily through the swelling of the polymer-protein matrix. The releasing kinetics of the proteins can be modulated through the addition of divalent cations, such as calcium. Notably, the polymer is not toxic to human dermal neonatal fibroblasts in vitro at RPS concentrations bellow 0.1 mg mL-1 . The results show that this polymer is a good candidate for the delivery of therapeutic macromolecules.
Collapse
Affiliation(s)
- José P Leite
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Rita Mota
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Faculdade de Ciências, Departamento de Biologia, Universidade do Porto, Rua do Campo Alegre, Edifício FC4, 4169-007, Porto, Portugal
| | - Joana Durão
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Sara C Neves
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,FEUP - Faculdade de Engenharia da Universidade do Porto, Departamento de Engenharia Metalúrgica e de Materiais, Rua Dr. Roberto Frias s/n, 4200-465, Porto, Portugal
| | - Cristina C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Paula Tamagnini
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Faculdade de Ciências, Departamento de Biologia, Universidade do Porto, Rua do Campo Alegre, Edifício FC4, 4169-007, Porto, Portugal
| | - Luís Gales
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| |
Collapse
|
35
|
Wu JL, Tian GX, Yu WJ, Jia GT, Sun TY, Gao ZQ. pH-Responsive Hyaluronic Acid-Based Mixed Micelles for the Hepatoma-Targeting Delivery of Doxorubicin. Int J Mol Sci 2016; 17:364. [PMID: 27043540 PMCID: PMC4848880 DOI: 10.3390/ijms17040364] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/28/2016] [Accepted: 03/07/2016] [Indexed: 02/07/2023] Open
Abstract
The tumor targetability and stimulus responsivity of drug delivery systems are crucial in cancer diagnosis and treatment. In this study, hepatoma-targeting mixed micelles composed of a hyaluronic acid-glycyrrhetinic acid conjugate and a hyaluronic acid-l-histidine conjugate (HA-GA/HA-His) were prepared through ultrasonic dispersion. The formation and characterization of the mixed micelles were confirmed via ¹H-NMR, particle size, and ζ potential measurements. The in vitro cellular uptake of the micelles was evaluated using human liver carcinoma (HepG2) cells. The antitumor effect of doxorubicin (DOX)-loaded micelles was investigated in vitro and in vivo. Results indicated that the DOX-loaded HA-GA/HA-His micelles showed a pH-dependent controlled release and were remarkably absorbed by HepG2 cells. Compared with free DOX, the DOX-loaded HA-GA/HA-His micelles showed a higher cytotoxicity to HepG2 cells. Moreover, the micelles effectively inhibited tumor growth in H22 cell-bearing mice. These results suggest that the HA-GA/HA-His mixed micelles are a good candidate for drug delivery in the prevention and treatment of hepatocarcinoma.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Cell Survival/drug effects
- Doxorubicin/administration & dosage
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Drug Carriers/chemistry
- Female
- Hep G2 Cells
- Histidine/chemistry
- Humans
- Hyaluronic Acid/chemistry
- Hydrogen-Ion Concentration
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Magnetic Resonance Spectroscopy
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Micelles
- Microscopy, Electron, Transmission
- Particle Size
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Jing-Liang Wu
- School of Bioscience and Technology, Weifang Medical University, Wei Fang 261053, Shandong, China.
| | - Gui-Xiang Tian
- School of Bioscience and Technology, Weifang Medical University, Wei Fang 261053, Shandong, China.
| | - Wen-Jing Yu
- School of Bioscience and Technology, Weifang Medical University, Wei Fang 261053, Shandong, China.
| | - Guang-Tao Jia
- School of Bioscience and Technology, Weifang Medical University, Wei Fang 261053, Shandong, China.
| | - Tong-Yi Sun
- School of Bioscience and Technology, Weifang Medical University, Wei Fang 261053, Shandong, China.
| | - Zhi-Qin Gao
- School of Bioscience and Technology, Weifang Medical University, Wei Fang 261053, Shandong, China.
| |
Collapse
|
36
|
Dag A, Callari M, Lu H, Stenzel MH. Modulating the cellular uptake of platinum drugs with glycopolymers. Polym Chem 2016. [DOI: 10.1039/c5py01579k] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The therapeutic potency of platinum-based anticancer drugs can be substantially improved through the use of fructose-coated nanocarrier systems to target cancer cells efficiently.
Collapse
Affiliation(s)
- Aydan Dag
- Centre for Advanced Macromolecular Design
- School of Chemistry
- The University of New South Wales
- Sydney
- Australia
| | - Manuela Callari
- Centre for Advanced Macromolecular Design
- School of Chemistry
- The University of New South Wales
- Sydney
- Australia
| | - Hongxu Lu
- Centre for Advanced Macromolecular Design
- School of Chemistry
- The University of New South Wales
- Sydney
- Australia
| | - Martina H. Stenzel
- Centre for Advanced Macromolecular Design
- School of Chemistry
- The University of New South Wales
- Sydney
- Australia
| |
Collapse
|
37
|
TANAKA T. Recent Advances in Glycopolymers Based on Protecting-Group-Free Synthesis. KOBUNSHI RONBUNSHU 2016. [DOI: 10.1295/koron.2016-0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tomonari TANAKA
- Department of Biobased Materials Science, Graduate School of Science and Technology, Kyoto Institute of Technology
| |
Collapse
|