1
|
Jourdain MA, Eyer J. Recent advances in liposomes and peptide-based therapeutics for glioblastoma treatment. J Control Release 2024; 376:732-752. [PMID: 39437968 DOI: 10.1016/j.jconrel.2024.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
In the context of glioblastoma treatment, the penetration of drugs is drastically limited by the blood-brain-barrier (BBB). Emerging therapies have focused on the field of therapeutic peptides for their excellent BBB targeting properties that promote a deep tumor penetration. Peptide-based strategies are also renowned for their abilities of driving cargo such as liposomal system allowing an active targeting of receptors overexpressed on GBM cells. This review provides a detailed description of the internalization mechanisms of specific GBM homing and penetrating peptides as well as the latest in vitro/in vivo studies of liposomes functionalized with them. The purpose of this review is to summarize a selection of promising pre-clinical results that demonstrate the advantages of this nanosystem, including an increase of tumor cell targeting, triggering drug accumulation and thus a strong antitumor effect. Aware of the early stage of these studies, many challenges need to be overcome to promote peptide-directed liposome at clinical level. In particular, the lack of suitable production, the difficulty to characterize the nanosystem and therapeutic competition leaded by antibodies.
Collapse
Affiliation(s)
- M-A Jourdain
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France.
| | - J Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|
2
|
Awuah WA, Ahluwalia A, Tan JK, Sanker V, Roy S, Ben-Jaafar A, Shah DM, Tenkorang PO, Aderinto N, Abdul-Rahman T, Atallah O, Alexiou A. Theranostics advances in the treatment and diagnosis of neurological and neurosurgical diseases. Arch Med Res 2024; 56:103085. [PMID: 39369666 DOI: 10.1016/j.arcmed.2024.103085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024]
Abstract
Theranostics represents a significant advance in the fields of neurology and neurosurgery, offering innovative approaches that combine the diagnosis and treatment of various neurological disorders. This innovation serves as a cornerstone of personalized medicine, where therapeutic strategies are closely integrated with diagnostic tools to enable precise and targeted interventions. Primary research results emphasize the profound impact of theranostics in Neuro Oncol. In this context, it has provided valuable insights into the complexity of the tumor microenvironment and mechanisms of resistance. In addition, in the field of neurodegenerative diseases (NDs), theranostics has facilitated the identification of distinct disease subtypes and novel therapeutic targets. It has also unravelled the intricate pathophysiology underlying conditions such as cerebrovascular disease (CVD) and epilepsy, setting the stage for more refined treatment approaches. As theranostics continues to evolve through ongoing research and refinement, its goals include further advancing the field of precision medicine, developing practical biomarkers for clinical use, and opening doors to new therapeutic opportunities. Nevertheless, the integration of these approaches into clinical settings presents challenges, including ethical considerations, the need for advanced data interpretation, standardization of procedures, and ensuring cost-effectiveness. Despite these obstacles, the promise of theranostics to significantly improve patient outcomes in the fields of neurology and neurosurgery remains a source of optimism for the future of healthcare.
Collapse
Affiliation(s)
| | - Arjun Ahluwalia
- School of Medicine, Queen's University Belfast, Belfast, United Kingdom
| | | | - Vivek Sanker
- Department of Neurosurgery, Stanford University, CA, USA
| | - Sakshi Roy
- School of Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Adam Ben-Jaafar
- University College Dublin, School of Medicine, Belfield, Dublin 4, Ireland
| | - Devansh Mitesh Shah
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | | | - Nicholas Aderinto
- Internal Medicine Department, LAUTECH Teaching Hospital, Ogbomoso, Nigeria
| | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Athanasios Alexiou
- University Centre for Research and Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research and Development, Funogen, Athens, Greece; Department of Research and Development, AFNP Med, Wien, Austria; Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia.
| |
Collapse
|
3
|
Yang H, Wu P, Wang T, Yu Y, Li J, Liu R, Ruan Q. Topical ophthalmic instillation of engineered hMSCs-derived exosomes: A novel non-invasive therapeutic strategy for ocular posterior-segment disorder. Biochem Biophys Res Commun 2024; 723:150212. [PMID: 38850812 DOI: 10.1016/j.bbrc.2024.150212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Due to the presence of protective mechanisms and blood-ocular barriers in the eye, drugs aimed at treating posterior segment ophthalmic disorder have to be administrated mostly through periocular or intravitreal injection. In the current study, we sought to investigate whether topical ophthalmic instillation of human mesenchymal stem cells (hMSCs)-derived exosomes can prevent and treat experimental autoimmune uveitis (EAU), a posterior segment ophthalmic disease induced in animals and considered a model of human autoimmune diseases of the eye. Our studies reveal that topical ophthalmic instillation of hMSCs-derived exosomes can effectively ameliorate EAU. More importantly, we demonstrate that exosomes modified by trans-activator of transcription peptide (TAT) were more effective than naive exosomes in penetrating ocular barrier and preventing/treating EAU. Taken together, these results indicate that topical ophthalmic instillation of TAT-peptide modified exosomes represents a novel non-invasive therapeutic strategy for posterior-segment ophthalmic disorders.
Collapse
Affiliation(s)
- Huiying Yang
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, 250000, China; Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
| | - Peipei Wu
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Ting Wang
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250021, China
| | - Yang Yu
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
| | - Jun Li
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Ruiling Liu
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China; Henan Key Laboratory of Engineering Antibody Medicine, Medical College of Henan University, Kaifeng, 475004, China.
| | - Qingguo Ruan
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China; Henan Key Laboratory of Engineering Antibody Medicine, Medical College of Henan University, Kaifeng, 475004, China.
| |
Collapse
|
4
|
Gan Y, Yu Y, Xu H, Piao H. Liposomal Nanomaterials: A Rising Star in Glioma Treatment. Int J Nanomedicine 2024; 19:6757-6776. [PMID: 38983132 PMCID: PMC11232959 DOI: 10.2147/ijn.s470478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/22/2024] [Indexed: 07/11/2024] Open
Abstract
Glioma is a primary malignant tumor in the central nervous system. In recent years, the treatment of glioma has developed rapidly, but the overall survival of glioma patients has not significantly improved. Due to the presence of the blood-brain barrier and intracranial tumor barrier, many drugs with good effects to cure glioma in vitro cannot be accurately transported to the corresponding lesions. In order to enable anti-tumor drugs to overcome the barriers and target glioma, nanodrug delivery systems have emerged recently. It is gratifying that liposomes, as a multifunctional nanodrug delivery carrier, which can be compatible with hydrophilic and hydrophobic drugs, easily functionalized by various targeted ligands, biodegradable, and hypoimmunogenic in vivo, has become a quality choice to solve the intractable problem of glioma medication. Therefore, we focused on the liposome nanodrug delivery system, and summarized its current research progress in glioma. Hopefully, this review may provide new ideas for the research and development of liposome-based nanomaterials for the clinical treatment of glioma.
Collapse
Affiliation(s)
- Yu Gan
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
| | - Yingying Yu
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
| | - Huizhe Xu
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
| |
Collapse
|
5
|
Wang X, Wan W, Lu J, Liu P. Inhalable FN-binding liposomes or liposome-exosome hybrid bionic vesicles encapsulated microparticles for enhanced pulmonary fibrosis therapy. Int J Pharm 2024; 656:124096. [PMID: 38583821 DOI: 10.1016/j.ijpharm.2024.124096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/09/2024]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive and irreversible interstitial lung disease that seriously threatens human life and health. Our previous study demonstrated the unique superiority of traditional Chinese medicine cryptotanshinone (CTS) combined with sustained pulmonary drug delivery for treating PF. In this study, we aimed to enhance the selectivity, targeting efficiency and sustained-release capability based on this delivery system. To this end, we developed and evaluated CTS-loaded modified liposomes-chitosan (CS) microspheres SM(CT-lipo) and liposome-exosome hybrid bionic vesicles-CS microspheres SM(LE). The prepared nano-in-micro particles system integrates the advantages of the carriers and complements each other. SM(CT-lipo) and SM(LE) achieved lung myofibroblast-specific targeting through CREKA peptide binding specifically to fibronectin (FN) and the homing effect of exosomes on parent cells, respectively, facilitating efficient delivery of anti-fibrosis drugs to lung lesions. Furthermore, compared with daily administration of conventional microspheres SM(NC) and positive control drug pirfenidone (PFD), inhaled administration of SM(CT-lipo) and SM(LE) every two days still attained similar efficacy, exhibiting excellent sustained drug release ability. In summary, our findings suggest that the developed SM(CT-lipo) and SM(LE) delivery strategies could achieve more accurate, efficient and safe therapy, providing novel insights into the treatment of chronic PF.
Collapse
Affiliation(s)
- Xiuhua Wang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei Wan
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jing Lu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Peiqing Liu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
6
|
Zhao C, Zhu X, Tan J, Mei C, Cai X, Kong F. Lipid-based nanoparticles to address the limitations of GBM therapy by overcoming the blood-brain barrier, targeting glioblastoma stem cells, and counteracting the immunosuppressive tumor microenvironment. Biomed Pharmacother 2024; 171:116113. [PMID: 38181717 DOI: 10.1016/j.biopha.2023.116113] [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: 10/20/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor, characterized by high heterogeneity, strong invasiveness, poor prognosis, and a low survival rate. A broad range of nanoparticles have been recently developed as drug delivery systems for GBM therapy owing to their inherent size effect and ability to cross the blood-brain barrier (BBB). Lipid-based nanoparticles (LBNPs), such as liposomes, solid lipid NPs (SLNs), and nano-structured lipid carriers (NLCs), have emerged as the most promising drug delivery system for the treatment of GBM because of their unique size, surface modification possibilities, and proven bio-safety. In this review, the main challenges of the current clinical treatment of GBM and the strategies on how novel LBNPs overcome them were explored. The application and progress of LBNP-based drug delivery systems in GBM chemotherapy, immunotherapy, and gene therapy in recent years were systematically reviewed, and the prospect of LBNPs for GBM treatment was discussed.
Collapse
Affiliation(s)
- Changhong Zhao
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China; Lantian Pharmaceuticals Co., Ltd, Hubei, China.
| | - Xinshu Zhu
- School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an 223005, China
| | - Jianmei Tan
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Chao Mei
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Xiang Cai
- Lantian Pharmaceuticals Co., Ltd, Hubei, China; School of Business, Hubei University of Science and Technology, China
| | - Fei Kong
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China; School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
7
|
Bataille Backer P, Adekiya TA, Kim Y, Reid TER, Thomas M, Adesina SK. Development of a Targeted SN-38-Conjugate for the Treatment of Glioblastoma. ACS OMEGA 2024; 9:2615-2628. [PMID: 38250376 PMCID: PMC10795035 DOI: 10.1021/acsomega.3c07486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024]
Abstract
Glioblastoma (GBM) is the most aggressive and fatal brain tumor, with approximately 10,000 people diagnosed every year in the United States alone. The typical survival period for individuals with glioblastoma ranges from 12 to 18 months, with significant recurrence rates. Common therapeutic modalities for brain tumors are chemotherapy and radiotherapy. The main challenges with chemotherapy for the treatment of glioblastoma are high toxicity, poor selectivity, and limited accumulation of therapeutic anticancer agents in brain tumors as a result of the presence of the blood-brain barrier. To overcome these challenges, researchers have explored strategies involving the combination of targeting peptides possessing a specific affinity for overexpressed cell-surface receptors with conventional chemotherapy agents via the prodrug approach. This approach results in the creation of peptide drug conjugates (PDCs), which facilitate traversal across the blood-brain barrier (BBB), enable preferential accumulation of chemotherapy within the neoplastic microenvironment, and selectively target cancerous cells. This approach increases accumulation in tumors, thereby improving therapeutic efficiency and minimizing toxicity. Leveraging the affinity of the HAIYPRH (T7) peptide for the transferrin receptor (TfR) overexpressed on the blood-brain barrier and glioma cells, a novel T7-SN-38 peptide drug conjugate was developed. The T7-SN-38 peptide drug conjugate demonstrates about a 2-fold reduction in glide score (binding affinity) compared to T7 while maintaining a comparable orientation within the TfR target site using Schrödinger-2022-3 Maestro 13.3 for ligand preparation and Glide SP-Peptide docking. Additionally, SN-38 extends into a solvent-accessible region, enhancing its susceptibility to protease hydrolysis at the cathepsin B (Cat B) cleavable site. The SN-38-ether-peptide drug conjugate displayed high stability in buffer at physiological pH, and cleavage of the conjugate to release free cytotoxic SN-38 was observed in the presence of exogenous cathepsin B. The synthesized peptide drug conjugate exhibited potent cytotoxic activities in cellular models of glioblastoma in vitro. In addition, blocking transferrin receptors using the free T7 peptide resulted in a notable inhibition of cytotoxicity of the conjugate, which was reversed when exogenous cathepsin B was added to cells. This work demonstrates the potential for targeted drug delivery to the brain in the treatment of glioblastoma using the transferrin receptor-targeted T7-SN-38 conjugate.
Collapse
Affiliation(s)
| | - Tayo Alex Adekiya
- Department
of Pharmaceutical Sciences, Howard University, Washington D.C. 20059, United States
| | - Yushin Kim
- Department
of Pharmaceutical Sciences, Concordia University
of Wisconsin, Mequon, Wisconsin 53097-2402, United States
| | - Terry-Elinor R. Reid
- Department
of Pharmaceutical Sciences, Concordia University
of Wisconsin, Mequon, Wisconsin 53097-2402, United States
| | - Michael Thomas
- Department
of Biology, Howard University, Washington D.C. 20059, United States
| | - Simeon K. Adesina
- Department
of Pharmaceutical Sciences, Howard University, Washington D.C. 20059, United States
| |
Collapse
|
8
|
Sonam Dongsar T, Tsering Dongsar T, Molugulu N, Annadurai S, Wahab S, Gupta N, Kesharwani P. Targeted therapy of breast tumor by PLGA-based nanostructures: The versatile function in doxorubicin delivery. ENVIRONMENTAL RESEARCH 2023; 233:116455. [PMID: 37356522 DOI: 10.1016/j.envres.2023.116455] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/15/2023] [Accepted: 06/17/2023] [Indexed: 06/27/2023]
Abstract
Breast carcinoma is a molecularly diverse illness, and it is among the most prominent and often reported malignancies in female across the globe. Surgical intervention, chemotherapy, immunotherapy, gene therapy, and endocrine treatment are among the currently viable treatment options for the carcinoma of breast. Chemotherapy is among the most prevalent cancer management strategy. Doxorubicin (DOX) widely employed as a cytostatic medication for the treatment of a variety of malignancies. Despite its widespread acceptance and excellent efficacy against an extensive line up of neoplasia, it has a variety of shortcomings that limit its therapeutic potential in the previously mentioned indications. Employment of nanoparticulate systems has come up as a unique chemo medication delivery strategy and are being considerably explored for the amelioration of breast carcinoma. Polylactic-co-glycolic acid (PLGA)-based nano systems are being utilized in a number of areas within the medical research and medication delivery constitutes one of the primary functions for PLGA given their inherent physiochemical attributes, including their aqueous solubility, biocompatibility, biodegradability, versatility in formulation, and limited toxicity. Herein along with the different application of PLGA-based nano formulations in cancer therapy, the present review intends to describe the various research investigations that have been conducted to enumerate the effectiveness of DOX-encapsulated PLGA nanoparticles (DOX-PLGA NPs) as a feasible treatment option for breast cancer.
Collapse
Affiliation(s)
- Tenzin Sonam Dongsar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Tenzin Tsering Dongsar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nagashekhara Molugulu
- School of Pharmacy, Monash University, Bandar Sunway, Jalan Lagoon Selatan, 47500, Malaysia
| | - Sivakumar Annadurai
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Neelima Gupta
- Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
9
|
Parrasia S, Szabò I, Zoratti M, Biasutto L. Peptides as Pharmacological Carriers to the Brain: Promises, Shortcomings and Challenges. Mol Pharm 2022; 19:3700-3729. [PMID: 36174227 DOI: 10.1021/acs.molpharmaceut.2c00523] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Central nervous system (CNS) diseases are among the most difficult to treat, mainly because the vast majority of the drugs fail to cross the blood-brain barrier (BBB) or to reach the brain at concentrations adequate to exert a pharmacological activity. The obstacle posed by the BBB has led to the in-depth study of strategies allowing the brain delivery of CNS-active drugs. Among the most promising strategies is the use of peptides addressed to the BBB. Peptides are versatile molecules that can be used to decorate nanoparticles or can be conjugated to drugs, with either a stable link or as pro-drugs. They have been used to deliver to the brain both small molecules and proteins, with applications in diverse therapeutic areas such as brain cancers, neurodegenerative diseases and imaging. Peptides can be generally classified as receptor-targeted, recognizing membrane proteins expressed by the BBB microvessels (e.g., Angiopep2, CDX, and iRGD), "cell-penetrating peptides" (CPPs; e.g. TAT47-57, SynB1/3, and Penetratin), undergoing transcytosis through unspecific mechanisms, or those exploiting a mixed approach. The advantages of peptides have been extensively pointed out, but so far few studies have focused on the potential negative aspects. Indeed, despite having a generally good safety profile, some peptide conjugates may display toxicological characteristics distinct from those of the peptide itself, causing for instance antigenicity, cardiovascular alterations or hemolysis. Other shortcomings are the often brief lifetime in vivo, caused by the presence of peptidases, the vulnerability to endosomal/lysosomal degradation, and the frequently still insufficient attainable increase of brain drug levels, which remain below the therapeutically useful concentrations. The aim of this review is to analyze not only the successful and promising aspects of the use of peptides in brain targeting but also the problems posed by this strategy for drug delivery.
Collapse
Affiliation(s)
- Sofia Parrasia
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| |
Collapse
|
10
|
Zhu Z, Zhai Y, Hao Y, Wang Q, Han F, Zheng W, Hong J, Cui L, Jin W, Ma S, Yang L, Cheng G. Specific anti-glioma targeted-delivery strategy of engineered small extracellular vesicles dual-functionalised by Angiopep-2 and TAT peptides. J Extracell Vesicles 2022; 11:e12255. [PMID: 35932288 PMCID: PMC9451528 DOI: 10.1002/jev2.12255] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/04/2022] [Accepted: 07/22/2022] [Indexed: 01/06/2023] Open
Abstract
Glioma is one of the primary malignant brain tumours in adults, with a poor prognosis. Pharmacological reagents targeting glioma are limited to achieve the desired therapeutic effect due to the presence of blood-brain barrier (BBB). Effectively crossing the BBB and specifically targeting to the brain tumour are the major challenge for the glioma treatments. Here, we demonstrate that the well-defined small extracellular vesicles (sEVs) with dual-targeting drug delivery and cell-penetrating functions, modified by Angiopep-2 and trans-activator of transcription peptides, enable efficient and specific chemotherapy for glioma. The high efficiency of engineered sEVs in targeting BBB and glioma was assessed in both monolayer culture cells and BBB model in vitro, respectively. The observed high targeting efficiency was re-validated in subcutaneous tumour and orthotopic glioma mice models. After loading the doxorubicin into dual-modified functional sEVs, this specific dual-targeting delivery system could cross the BBB, reach the glioma, and penetrate the tumour. Such a mode of drug delivery significantly improved more than 2-fold survival time of glioma mice with very few side effects. In conclusion, utilization of the dual-modified sEVs represents a unique and efficient strategy for drug delivery, holding great promise for the treatments of central nervous system diseases.
Collapse
Affiliation(s)
- Zhanchi Zhu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Yuanxin Zhai
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Ying Hao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China.,Guangdong Institute of Semiconductor Micro-Nano Manufacturing Technology, Guangdong, China
| | - Quanwei Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Fang Han
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Wenlong Zheng
- Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou, China
| | - Jing Hong
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Leisha Cui
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Wei Jin
- Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Sancheng Ma
- Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou, China
| | - Lingyan Yang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Guosheng Cheng
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou, China.,Guangdong Institute of Semiconductor Micro-Nano Manufacturing Technology, Guangdong, China
| |
Collapse
|
11
|
Taghipour YD, Zarebkohan A, Salehi R, Rahimi F, Torchilin VP, Hamblin MR, Seifalian A. An update on dual targeting strategy for cancer treatment. J Control Release 2022; 349:67-96. [PMID: 35779656 DOI: 10.1016/j.jconrel.2022.06.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/04/2022] [Accepted: 06/24/2022] [Indexed: 12/18/2022]
Abstract
The key issue in the treatment of solid tumors is the lack of efficient strategies for the targeted delivery and accumulation of therapeutic cargoes in the tumor microenvironment (TME). Targeting approaches are designed for more efficient delivery of therapeutic agents to cancer cells while minimizing drug toxicity to normal cells and off-targeting effects, while maximizing the eradication of cancer cells. The highly complicated interrelationship between the physicochemical properties of nanoparticles, and the physiological and pathological barriers that are required to cross, dictates the need for the success of targeting strategies. Dual targeting is an approach that uses both purely biological strategies and physicochemical responsive smart delivery strategies to increase the accumulation of nanoparticles within the TME and improve targeting efficiency towards cancer cells. In both approaches, either one single ligand is used for targeting a single receptor on different cells, or two different ligands for targeting two different receptors on the same or different cells. Smart delivery strategies are able to respond to triggers that are typical of specific disease sites, such as pH, certain specific enzymes, or redox conditions. These strategies are expected to lead to more precise targeting and better accumulation of nano-therapeutics. This review describes the classification and principles of dual targeting approaches and critically reviews the efficiency of dual targeting strategies, and the rationale behind the choice of ligands. We focus on new approaches for smart drug delivery in which synthetic and/or biological moieties are attached to nanoparticles by TME-specific responsive linkers and advanced camouflaged nanoparticles.
Collapse
Affiliation(s)
- Yasamin Davatgaran Taghipour
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Fariborz Rahimi
- Department of Electrical Engineering, University of Bonab, Bonab, Iran
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine and Department of Chemical Engineering, Northeastern University, Boston, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, South Africa
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
12
|
Hou Q, Zhu L, Wang L, Liu X, Xiao F, Xie Y, Zheng W, Jiang X. Screening on-chip fabricated nanoparticles for penetrating the blood-brain barrier. NANOSCALE 2022; 14:3234-3241. [PMID: 35156984 DOI: 10.1039/d1nr05825h] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The inability of drugs to cross the blood-brain barrier (BBB) makes it difficult to treat diseases in the central nervous system. It is known that peptides with or without specific receptors on the BBB showed different or even controversial neuron targeting capability in different reports. So, it is necessary to clarify how these peptides work as targeting molecules in the central nervous system. Herein, we evaluate and compare the performance of 6 kinds of peptides with (T7, D-T7, and GSH) or without (TGN, CGN, and TAT) BBB-specific receptors by conjugating these peptides on lipids to serve as a shell to encapsulate a core of PLGA and lamotrigine to form nanoparticles for targeted epilepsy therapy. In vitro assay shows that the TAT-modified nanoparticles show the highest internalization efficacy in the BBB model cell line bEnd·3 cells and hippocampal neurons. By contrast, experiments in mice show that the D-T7-modified nanoparticles have the highest brain targeting and epilepsy therapeutic efficiency. Thus, our experiments uncover the different performances of the 6 peptides at different levels (in vitro and in vivo), which is insightful for developing novel delivery systems for treating diseases in the central nervous system.
Collapse
Affiliation(s)
- Qinghong Hou
- Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, P. R. China.
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong 518055, P. R. China.
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, P. R. China.
| | - Lina Zhu
- Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, P. R. China.
| | - Le Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong 518055, P. R. China.
| | - Xiaoyan Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong 518055, P. R. China.
| | - Feng Xiao
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong 518055, P. R. China.
| | - Yangzhouyun Xie
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong 518055, P. R. China.
| | - Wenfu Zheng
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, P. R. China.
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong 518055, P. R. China.
| |
Collapse
|
13
|
Poustforoosh A, Nematollahi MH, Hashemipour H, Pardakhty A. Recent advances in Bio-conjugated nanocarriers for crossing the Blood-Brain Barrier in (pre-)clinical studies with an emphasis on vesicles. J Control Release 2022; 343:777-797. [DOI: 10.1016/j.jconrel.2022.02.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022]
|
14
|
Construction and characterization of a novel Tenofovir-loaded PEGylated niosome conjugated with TAT peptide for evaluation of its cytotoxicity and anti-HIV effects. ADV POWDER TECHNOL 2021. [DOI: 10.1016/j.apt.2021.05.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
15
|
Systemic metastasis-targeted nanotherapeutic reinforces tumor surgical resection and chemotherapy. Nat Commun 2021; 12:3187. [PMID: 34045459 PMCID: PMC8160269 DOI: 10.1038/s41467-021-23466-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 04/21/2021] [Indexed: 01/26/2023] Open
Abstract
Failure of conventional clinical therapies such as tumor resection and chemotherapy are mainly due to the ineffective control of tumor metastasis. Metastasis consists of three steps: (i) tumor cells extravasate from the primary sites into the circulation system via epithelial-mesenchymal transition (EMT), (ii) the circulating tumor cells (CTCs) form “micro-thrombi” with platelets to evade the immune surveillance in circulation, and (iii) the CTCs colonize in the pre-metastatic niche. Here, we design a systemic metastasis-targeted nanotherapeutic (H@CaPP) composed of an anti-inflammatory agent, piceatannol, and an anti-thrombotic agent, low molecular weight heparin, to hinder the multiple steps of tumor metastasis. H@CaPP is found efficiently impeded EMT, inhibited the formation of “micro-thrombi”, and prevented the development of pre-metastatic niche. When combined with surgical resection or chemotherapy, H@CaPP efficiently inhibits tumor metastasis and prolonged overall survival of tumor-bearing mice. Collectively, we provide a simple and effective systemic metastasis-targeted nanotherapeutic for combating tumor metastasis. Failure of conventional clinical therapies such as tumor resection and chemotherapy are mainly due to the ineffective control of tumor metastasis. Here, the authors show that a systemic metastasis-targeted nanotherapeutic may offer a powerful adjunct therapy for suppressing tumor metastasis.
Collapse
|
16
|
Izci M, Maksoudian C, Manshian BB, Soenen SJ. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem Rev 2021; 121:1746-1803. [PMID: 33445874 PMCID: PMC7883342 DOI: 10.1021/acs.chemrev.0c00779] [Citation(s) in RCA: 216] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 02/08/2023]
Abstract
Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational
Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| |
Collapse
|
17
|
Peptide-Based Nanomaterials for Tumor Immunotherapy. Molecules 2020; 26:molecules26010132. [PMID: 33396754 PMCID: PMC7796410 DOI: 10.3390/molecules26010132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022] Open
Abstract
With the increasing understanding of tumor immune circulation mechanisms, tumor immunotherapy including immune checkpoint blockade has become a research hotspot, which requires the development of more accurate and more efficient drugs with fewer side effects. In line with this requirement, peptides with good biocompatibility, targeting, and specificity become favorable theranostic reagents, and a series of promising candidates for tumor immunotherapy based on peptides have been developed. Additionally, the advantages of nanomaterials as drug carriers such as higher affinity have been demonstrated, providing possibilities of combination therapy. In this review, we summarize the development of peptide-based nanomaterials in tumor immunotherapy from the two aspects of functionalization and self-assembly. Furthermore, new methods for peptide screening, especially machine-learning-related strategies, is also a topic we were interested in, as this forms the basis for the construction of peptide-based platforms. Peptides provide broad prospects for tumor immunotherapy and we hope that this summary can provide insight into possible avenues for future exploration.
Collapse
|
18
|
Alfaifi AA, Heyder RS, Bielski ER, Almuqbil RM, Kavdia M, Gerk PM, da Rocha SRP. Megalin-targeting liposomes for placental drug delivery. J Control Release 2020; 324:366-378. [PMID: 32461116 PMCID: PMC8247794 DOI: 10.1016/j.jconrel.2020.05.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/09/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022]
Abstract
Every year, complications during pregnancy affect more than 26 million women. Some of those diseases are associated with significant morbidity and mortality, as is the case of preeclampsia, the main cause of maternal deaths globally. The ability to improve the delivery of drugs to the placenta upon administration to the mother may offer new opportunities in the treatment of diseases of pregnancy. The objective of this study was to develop megalin-targeting liposome nanocarriers for placental drug delivery. Megalin is a transmembrane protein involved in clathrin-mediated endocytic processes, and is expressed in the syncytiotrophoblast (SynT), an epithelial layer at maternal-fetal interface. Targeting megalin thus offers an opportunity for the liposomes to hitchhike into the SynT, thus enriching the concentration of any associated therapeutic cargo in the placental tissue. PEGylated (2 KDa) lipids were modified with gentamicin (GM), a substrate to megalin receptors as we have shown in earlier studies, and used to prepare placental-targeting liposomes. The ability of the targeting liposomes to enhance accumulation of a fluorescence probe was assessed in an in vivo placental model - timed-pregnant Balb/c mice at gestational day (GD) 18.5. The targeting liposomes containing 10 mol% GM-modified lipids increased the accumulation of the conjugated fluorescence probe in the placenta with a total accumulation of 2.8% of the initial dose, which corresponds to a 94 fold increase in accumulation compared to the free probe (p < .0001), and 2-4 fold accumulation compared to the non-targeting control liposomes (p < .0001), as measured by both tissue extraction assay and ex vivo imaging. Furthermore, confocal images of placental SynT cross-sections show a 3-fold increase of the targeting liposomes compared with the non-targeting liposomes. The rate and extent of uptake of a fluorescent probe encapsulated within targeting liposomes was also probed in an in vitro model of the human placental barrier (polarized BeWo monolayers) using flow cytometry. Targeting liposomes containing 5 mol% GM-modified lipids enhanced the uptake of the probe by 1.5 fold compared to the non-targeting control. An increase to 10 mol% of the modified lipid resulted in further enhancement in uptake, which was 2 fold greater compared to control. In a competition assay, inhibition of the megalin receptors resulted in a significant reduction in uptake of the fluorescence probe encapsulated in GM-modified liposomes compared to the uptake without free inhibitor (p < .0001), implicating the involvement of megalin receptor in the internalization of the liposomes. Taken together, these results demonstrate that megalin-targeted liposomes may offer an opportunity to enhance the delivery of therapeutics to the placenta for the treatment of diseases of pregnancy.
Collapse
Affiliation(s)
- Ali A Alfaifi
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, United States of America; Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Rodrigo S Heyder
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Elizabeth R Bielski
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Rashed M Almuqbil
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, United States of America
| | - Phillip M Gerk
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Sandro R P da Rocha
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States of America.
| |
Collapse
|
19
|
Darrigues E, Nima ZA, Griffin RJ, Anderson JM, Biris AS, Rodriguez A. 3D cultures for modeling nanomaterial-based photothermal therapy. NANOSCALE HORIZONS 2020; 5:400-430. [PMID: 32118219 DOI: 10.1039/c9nh00628a] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Photothermal therapy (PTT) is one of the most promising techniques for cancer tumor ablation. Nanoparticles are increasingly being investigated for use with PTT and can serve as theranostic agents. Based on the ability of near-infrared nano-photo-absorbers to generate heat under laser irradiation, PTT could prove advantageous in certain situations over more classical cancer therapies. To analyze the efficacy of nanoparticle-based PTT, preclinical in vitro studies typically use 2D cultures, but this method cannot completely mimic the complex tumor organization, bioactivity, and physiology that all control the complex penetration depth, biodistribution, and tissue diffusion parameters of nanomaterials in vivo. To fill this knowledge gap, 3D culture systems have been explored for PTT analysis. These models provide more realistic microenvironments that allow spatiotemporal oxygen gradients and cancer cell adaptations to be considered. This review highlights the work that has been done to advance 3D models for cancer microenvironment modeling, specifically in the context of advanced, functionalized nanoparticle-directed PTT.
Collapse
Affiliation(s)
- Emilie Darrigues
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S University Avenue, Little Rock, AR 72204, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Vale N, Alves C, Sharma V, Lázaro DF, Silva S, Gomes P, Outeiro TF. A new MAP-Rasagiline conjugate reduces α-synuclein inclusion formation in a cell model. Pharmacol Rep 2020; 72:456-464. [PMID: 32048262 DOI: 10.1007/s43440-019-00032-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/05/2019] [Accepted: 11/18/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disease of the elderly. Current therapies are only symptomatic, and have no disease-modifying effect. Therefore, disease progresses continuously over time, presenting with both motor and non-motor features. The precise molecular basis for PD is still elusive, but the aggregation of the protein alpha-synuclein (α-syn) is a key pathological hallmark of the disease and is, therefore, a major focus of current research. Considering the intrinsic properties of cell-penetrating peptides (CPPs) for mediating drug delivery of neurotherapeutics across the blood brain barrier (BBB), these might open novel opportunities for the development of new solutions for the treatment of brain-related aspects of PD and other neurodegenerative disorders. METHODS Here, we synthesized solid-phase CPPs using an amphipathic model peptide (MAP) conjugated with the drug Rasagiline (RAS), which we named RAS-MAP, and evaluated its effect on α-syn inclusion formation in a human cell-based model of synucleinopathy. RESULTS We found that treatment with RAS-MAP at low concentrations (1-3 µM) reduced α-syn aggregation in cells. CONCLUSIONS For the first time, we report that conjugation of a current drug used in the therapy of PD with CPP reduces α-syn aggregation, which might prove beneficial in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Nuno Vale
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal. .,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal. .,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal. .,Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| | - Cláudia Alves
- Department of Chemistry and Biochemistry, Faculty of Sciences, LAQV/REQUIMTE, University of Porto, Rua do Campo Alegre, 687, 4169-007, Porto, Portugal
| | - Vaishali Sharma
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| | - Diana F Lázaro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| | - Sara Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Paula Gomes
- Department of Chemistry and Biochemistry, Faculty of Sciences, LAQV/REQUIMTE, University of Porto, Rua do Campo Alegre, 687, 4169-007, Porto, Portugal
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
21
|
Araste F, Abnous K, Hashemi M, Dehshahri A, Detampel P, Alibolandi M, Ramezani M. Na +/K + ATPase-targeted delivery to metastatic breast cancer models. Eur J Pharm Sci 2019; 143:105207. [PMID: 31870814 DOI: 10.1016/j.ejps.2019.105207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/08/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
In this study, we reported doxorubicin (DOX)-encapsulated nanoparticles (NPs) formulated with biocompatible and biodegradable poly (lactic-co-glycolic acid) (PLGA) and modified with a 13-amino acid peptide (S3) against sodium/potassium (Na+/K+)-ATPase pump alpha subunit to investigate its potential as antitumor agent. The morphological properties and size dispersity of the prepared nanoparticles were evaluated using scanning electron microscope (SEM) and dynamic light scattering (DLS). The encapsulation efficiency and in vitro release during 7 days were evaluated. Comparative in vitro cytotoxicity experiments demonstrated that the S3-conjugated nanoparticles (S3-PLGA-DOX NPs) had higher antiproliferative activity. Flow cytometry analysis confirmed the enhanced cellular uptake of S3-PLGA-DOX NPs in comparison with PLGA-DOX. In vivo study in 4T1 tumor-bearing BALB/C mice revealed that the S3-functionalized DOX-loaded NPs improved antitumor activity and survival rate of 4T1 tumor bearing mice. In this regard, conjugation of S3 peptide to the surface of DOX-loaded PLGA NPs provides site-specific delivery of DOX, inhibits 4T1 tumor growth in vivo and significantly decreases systemic toxicity. The obtained results suggested that the new (Na+/K+)-ATPase pump-targeted PLGA NPs as a target-selective delivery system for DOX has great potential for the treatment of breast cancer.
Collapse
Affiliation(s)
- Fatemeh Araste
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pascal Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel, Switzerland
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
22
|
Johnsen KB, Burkhart A, Thomsen LB, Andresen TL, Moos T. Targeting the transferrin receptor for brain drug delivery. Prog Neurobiol 2019; 181:101665. [DOI: 10.1016/j.pneurobio.2019.101665] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/10/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
|
23
|
Xie J, Shen Z, Anraku Y, Kataoka K, Chen X. Nanomaterial-based blood-brain-barrier (BBB) crossing strategies. Biomaterials 2019; 224:119491. [PMID: 31546096 DOI: 10.1016/j.biomaterials.2019.119491] [Citation(s) in RCA: 289] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/31/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022]
Abstract
Increasing attention has been paid to the diseases of central nervous system (CNS). The penetration efficiency of most CNS drugs into the brain parenchyma is rather limited due to the existence of blood-brain barrier (BBB). Thus, BBB crossing for drug delivery to CNS remains a significant challenge in the development of neurological therapeutics. Because of the advantageous properties (e.g., relatively high drug loading content, controllable drug release, excellent passive and active targeting, good stability, biodegradability, biocompatibility, and low toxicity), nanomaterials with BBB-crossability have been widely developed for the treatment of CNS diseases. This review summarizes the current understanding of the physiological structure of BBB, and provides various nanomaterial-based BBB-crossing strategies for brain delivery of theranostic agents, including intranasal delivery, temporary disruption of BBB, local delivery, cell penetrating peptide (CPP) mediated BBB-crossing, receptor mediated BBB-crossing, shuttle peptide mediated BBB-crossing, and cells mediated BBB-crossing. Clinicians, biologists, material scientists and chemists are expected to be interested in this review.
Collapse
Affiliation(s)
- Jinbing Xie
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China; Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Zheyu Shen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Yasutaka Anraku
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan; Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Shi D, Mi G, Shen Y, Webster TJ. Glioma-targeted dual functionalized thermosensitive Ferri-liposomes for drug delivery through an in vitro blood-brain barrier. NANOSCALE 2019; 11:15057-15071. [PMID: 31369016 DOI: 10.1039/c9nr03931g] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
To date, the delivery of therapeutic agents for malignant brain tumors (such as glioblastoma multiforme (GBM)) remains a significant obstacle due to the existence of the blood-brain barrier (BBB). A multitude of delivery systems (hydrogels, micelles, polymeric nanoparticles, etc.) have been proposed, yet many of them exhibit limited tumor-specific inhibition effects. Herein, a drug-encapsulated dual-functionalized thermosensitive liposomal system (DOX@P1NS/TNC-FeLP) was developed for targeted delivery across the BBB. Specifically, a GBM-specific cell-penetrating peptide (P1NS) and an anti-GBM antibody (TN-C) were conjugated onto the liposome surface for targeted delivery. In addition, superparamagnetic iron oxide nanoparticles (SPIONs) and doxorubicin (DOX) were co-loaded inside the liposomes to achieve thermo-triggered drug release when applying an alternating magnetic field (AMF). Results demonstrated that P1NS/TNC-FeLPs readily transported across an in vitro BBB model and displayed a thermo-responsive and GBM-specific cellular uptake as well as drug release profile. Additionally, results from immunofluorescent (IF) staining and RT-qPCR further demonstrated that DOX@P1NS/TNC-FeLPs specifically entered U-87 human GBM cells and suppressed tumor cell proliferation without causing any significant impact on healthy brain cell function. As such, the novel DOX@P1NS/TNC-FeLPs presented potent and precise anti-GBM capability and, therefore, are suggested here for the first time as a promising DDS to deliver therapeutic agents across the BBB for GBM treatment.
Collapse
Affiliation(s)
- Di Shi
- Chemical Engineering Department, Northeastern University, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
25
|
Yuan J, Zeng C, Cao W, Zhou X, Pan Y, Xie Y, Zhang Y, Yang Q, Wang S. Bufalin-Loaded PEGylated Liposomes: Antitumor Efficacy, Acute Toxicity, and Tissue Distribution. NANOSCALE RESEARCH LETTERS 2019; 14:223. [PMID: 31278603 PMCID: PMC6611856 DOI: 10.1186/s11671-019-3057-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 06/19/2019] [Indexed: 05/08/2023]
Abstract
Bufalin, derived from Venenum Bufonis, exerts antitumor effects but has low bioavailability and adverse effects when administered as a single agent. The purpose of this study was to evaluate the physical and chemical properties, antitumor efficacy, general pharmacology, acute toxicity, and tissue distribution profile of bufalin-loaded PEGylated liposomes (BF/PEG-LP), which were prepared in a previous study. To evaluate the safety of the preparation, a red blood cell hemolysis test was performed, which indicated that the hemolysis rate of BF/PEG-LP was significantly lower than that of bufalin alone. Cell viability assay revealed that the blank liposomes were nontoxic. In an in vitro experiment, BF/PEG-LP dose-dependently induced the apoptosis of HepG2, HCT116, A549, and U251 cancer cells, with half-maximal inhibitory concentration (IC50) values of 21.40 ± 2.39, 21.00 ± 3.34, 43.39 ± 6.43, and 31.14 ± 2.58 ng/mL, respectively, at 24 h. Tumor xenograft experiments in nude mice showed that BF/PEG-LP significantly inhibited the growth of U251 cells. Pharmacological evaluation revealed that BF/PEG-LP impacted the general behavior, independent activities, and coordination of mice after a week of administration compared with those of mice in the control group. In an acute toxicity test, the median lethal concentration (LD50) of BF and BF/PEG-LP in mice was 0.156 and 3.03 mg/kg, respectively. Tissue distribution profiles showed that the BF concentration in brain tissue was 20% higher, whereas that in heart tissue was 30% lower when BF/PEG-LP was administered to mice compared with BF. Thus, BF/PEG-LP exhibited lower hemolysis and cytotoxicity and improved pharmacokinetic and antitumor properties compared with bufalin alone, indicating its potential for future pharmacological application, particularly for glioma treatment.
Collapse
Affiliation(s)
- Jiani Yuan
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Cheng Zeng
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Wei Cao
- Shannxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A & F University, Yangling, China
| | - Xuanxuan Zhou
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Yang Pan
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Yanhua Xie
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Yifang Zhang
- Shaanxi Pharmaceutical Development Center, Xi’an, China
| | - Qian Yang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Siwang Wang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| |
Collapse
|
26
|
Kim J, Ahn SI, Kim Y. Nanotherapeutics Engineered to Cross the Blood-Brain Barrier for Advanced Drug Delivery to the Central Nervous System. J IND ENG CHEM 2019; 73:8-18. [PMID: 31588177 PMCID: PMC6777578 DOI: 10.1016/j.jiec.2019.01.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Drug delivery to the brain remains challenging mainly due to the blood-brain barrier (BBB) that regulates the entrance of substances to the brain. Advances in nanotechnology have enabled the engineering of nanomedicines for biomedical applications including enhanced drug delivery into the brain. In this review, we describe strategies of nanomedicines engineered to traverse the BBB and deliver therapeutic molecules to target brain sites. We highlight the representative applications with materials including polymers, lipids, and inorganic elements for brain drug delivery. We finalize this review with the current challenges and future perspective of nanotherapeutics for advanced drug delivery to the brain.
Collapse
Affiliation(s)
- Jinhwan Kim
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Institute for Electronics and Nanotechnology (IEN), Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Song Ih Ahn
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Institute for Electronics and Nanotechnology (IEN), Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - YongTae Kim
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Institute for Electronics and Nanotechnology (IEN), Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
27
|
Dang Y, An C, Li Y, Han D, Liu X, Zhang F, Xu Y, Zhong H, Karim Khan MK, Zou F, Sun X. Neutrophil-mediated and low density lipoprotein receptor-mediated dual-targeting nanoformulation enhances brain accumulation of scutellarin and exerts neuroprotective effects against ischemic stroke. RSC Adv 2019; 9:1299-1318. [PMID: 35518053 PMCID: PMC9059646 DOI: 10.1039/c8ra06688d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/05/2018] [Indexed: 11/21/2022] Open
Abstract
Delivery of poorly permeable drugs across the blood-brain barrier (BBB) is a great challenge in the treatment of ischemic stroke.
Collapse
|
28
|
Large DE, Soucy JR, Hebert J, Auguste DT. Advances in Receptor-Mediated, Tumor-Targeted Drug Delivery. ADVANCED THERAPEUTICS 2019; 2:1800091. [PMID: 38699509 PMCID: PMC11064891 DOI: 10.1002/adtp.201800091] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Indexed: 02/06/2023]
Abstract
Receptor-mediated drug delivery presents an opportunity to enhance therapeutic efficiency by accumulating drug within the tissue of interest and reducing undesired, off-target effects. In cancer, receptor overexpression is a platform for binding and inhibiting pathways that shape biodistribution, toxicity, cell binding and uptake, and therapeutic function. This review will identify tumor-targeted drug delivery vehicles and receptors that show promise for clinical translation based on quantitative in vitro and in vivo data. The authors describe the rationale to engineer a targeted drug delivery vehicle based on the ligand, chemical conjugation method, and type of drug delivery vehicle. Recent advances in multivalent targeting and ligand organization on tumor accumulation are discussed. Revolutionizing receptor-mediated drug delivery may be leveraged in the therapeutic delivery of chemotherapy, gene editing tools, and epigenetic drugs.
Collapse
Affiliation(s)
- Danielle E Large
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Jonathan R Soucy
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Jacob Hebert
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Debra T Auguste
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| |
Collapse
|
29
|
Accardo A, Mannucci S, Nicolato E, Vurro F, Diaferia C, Bontempi P, Marzola P, Morelli G. Easy formulation of liposomal doxorubicin modified with a bombesin peptide analogue for selective targeting of GRP receptors overexpressed by cancer cells. Drug Deliv Transl Res 2018; 9:215-226. [DOI: 10.1007/s13346-018-00606-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
30
|
Mendes M, Sousa JJ, Pais A, Vitorino C. Targeted Theranostic Nanoparticles for Brain Tumor Treatment. Pharmaceutics 2018; 10:E181. [PMID: 30304861 PMCID: PMC6321593 DOI: 10.3390/pharmaceutics10040181] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/21/2018] [Accepted: 09/27/2018] [Indexed: 12/13/2022] Open
Abstract
The poor prognosis and rapid recurrence of glioblastoma (GB) are associated to its fast-growing process and invasive nature, which make difficult the complete removal of the cancer infiltrated tissues. Additionally, GB heterogeneity within and between patients demands a patient-focused method of treatment. Thus, the implementation of nanotechnology is an attractive approach considering all anatomic issues of GB, since it will potentially improve brain drug distribution, due to the interaction between the blood⁻brain barrier and nanoparticles (NPs). In recent years, theranostic techniques have also been proposed and regarded as promising. NPs are advantageous for this application, due to their respective size, easy surface modification and versatility to integrate multiple functional components in one system. The design of nanoparticles focused on therapeutic and diagnostic applications has increased exponentially for the treatment of cancer. This dual approach helps to understand the location of the tumor tissue, the biodistribution of nanoparticles, the progress and efficacy of the treatment, and is highly useful for personalized medicine-based therapeutic interventions. To improve theranostic approaches, different active strategies can be used to modulate the surface of the nanotheranostic particle, including surface markers, proteins, drugs or genes, and take advantage of the characteristics of the microenvironment using stimuli responsive triggers. This review focuses on the different strategies to improve the GB treatment, describing some cell surface markers and their ligands, and reports some strategies, and their efficacy, used in the current research.
Collapse
Affiliation(s)
- Maria Mendes
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
| | - João José Sousa
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- LAQV, REQUIMTE, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal.
| | - Alberto Pais
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- LAQV, REQUIMTE, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal.
| |
Collapse
|
31
|
Ahlschwede KM, Curran GL, Rosenberg JT, Grant SC, Sarkar G, Jenkins RB, Ramakrishnan S, Poduslo JF, Kandimalla KK. Cationic carrier peptide enhances cerebrovascular targeting of nanoparticles in Alzheimer's disease brain. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 16:258-266. [PMID: 30300748 DOI: 10.1016/j.nano.2018.09.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/02/2018] [Accepted: 09/13/2018] [Indexed: 11/18/2022]
Abstract
Accumulation of amyloid beta (Aβ) peptides in the cerebral vasculature, referred to as cerebral amyloid angiopathy (CAA), is widely observed in Alzheimer's disease (AD) brain and was shown to accelerate cognitive decline. There is no effective method for detecting cerebrovascular amyloid (CVA) and treat CAA. The targeted nanoparticles developed in this study effectively migrated from the blood flow to the vascular endothelium as determined by using quartz crystal microbalance with dissipation monitoring (QCM-D) technology. We also improved the stability, and blood-brain barrier (BBB) transcytosis of targeted nanoparticles by coating them with a cationic BBB penetrating peptide (K16ApoE). The K16ApoE-Targeted nanoparticles demonstrated specific targeting of vasculotropic DutchAβ40 peptide accumulated in the cerebral vasculature. Moreover, K16ApoE-Targeted nanoparticles demonstrated significantly greater uptake into brain and provided specific MRI contrast to detect brain amyloid plaques.
Collapse
Affiliation(s)
- Kristen M Ahlschwede
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA; Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Geoffry L Curran
- Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jens T Rosenberg
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA; Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Samuel C Grant
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA; Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Gobinda Sarkar
- Department of Experimental Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Robert B Jenkins
- Department of Experimental Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Subramanian Ramakrishnan
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA; Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Joseph F Poduslo
- Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA; Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
32
|
Mészáros M, Porkoláb G, Kiss L, Pilbat AM, Kóta Z, Kupihár Z, Kéri A, Galbács G, Siklós L, Tóth A, Fülöp L, Csete M, Sipos Á, Hülper P, Sipos P, Páli T, Rákhely G, Szabó-Révész P, Deli MA, Veszelka S. Niosomes decorated with dual ligands targeting brain endothelial transporters increase cargo penetration across the blood-brain barrier. Eur J Pharm Sci 2018; 123:228-240. [PMID: 30031862 DOI: 10.1016/j.ejps.2018.07.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/13/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022]
Abstract
Nanoparticles targeting transporters of the blood-brain barrier (BBB) are promising candidates to increase the brain penetration of biopharmacons. Solute carriers (SLC) are expressed at high levels in brain endothelial cells and show a specific pattern at the BBB. The aim of our study was to test glutathione and ligands of SLC transporters as single or dual BBB targeting molecules for nanovesicles. High mRNA expression levels for hexose and neutral amino acid transporting SLCs were found in isolated rat brain microvessels and our rat primary cell based co-culture BBB model. Niosomes were derivatized with glutathione and SLC ligands glucopyranose and alanine. Serum albumin complexed with Evans blue (67 kDa), which has a very low BBB penetration, was selected as a cargo. The presence of targeting ligands on niosomes, especially dual labeling, increased the uptake of the cargo molecule in cultured brain endothelial cells. This cellular uptake was temperature dependent and could be decreased with a metabolic inhibitor and endocytosis blockers filipin and cytochalasin D. Making the negative surface charge of brain endothelial cells more positive with a cationic lipid or digesting the glycocalyx with neuraminidase elevated the uptake of the cargo after treatment with targeted nanocarriers. Treatment with niosomes increased plasma membrane fluidity, suggesting the fusion of nanovesicles with endothelial cell membranes. Targeting ligands elevated the permeability of the cargo across the BBB in the culture model and in mice, and dual-ligand decoration of niosomes was more effective than single ligand labeling. Our data indicate that dual labeling with ligands of multiple SLC transporters can potentially be exploited for BBB targeting of nanoparticles.
Collapse
Affiliation(s)
- Mária Mészáros
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Doctoral School in Theoretical Medicine, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Gergő Porkoláb
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Foundation for the Future of Biomedical Sciences in Szeged, Pálfy u. 52/d, H-6725 Szeged, Hungary
| | - Lóránd Kiss
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Ana-Maria Pilbat
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Zoltán Kóta
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Zoltán Kupihár
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Albert Kéri
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Informatics, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Gábor Galbács
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Informatics, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - László Siklós
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - András Tóth
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary, Közép fasor 52, H-6726 Szeged, Hungary
| | - Lívia Fülöp
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Mária Csete
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary
| | - Áron Sipos
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Petra Hülper
- Department of Pediatrics I, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Péter Sipos
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Tibor Páli
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Gábor Rákhely
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary, Közép fasor 52, H-6726 Szeged, Hungary
| | - Piroska Szabó-Révész
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary.
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary.
| |
Collapse
|
33
|
Xu G, Chen Y, Shan R, Wu X, Chen L. Transferrin and tocopheryl-polyethylene glycol-succinate dual ligands decorated, cisplatin loaded nano-sized system for the treatment of lung cancer. Biomed Pharmacother 2018; 99:354-362. [DOI: 10.1016/j.biopha.2018.01.062] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/05/2018] [Accepted: 01/11/2018] [Indexed: 12/14/2022] Open
|
34
|
Lu H, Stenzel MH. Multicellular Tumor Spheroids (MCTS) as a 3D In Vitro Evaluation Tool of Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1702858. [PMID: 29450963 DOI: 10.1002/smll.201702858] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/13/2017] [Indexed: 05/23/2023]
Abstract
Multicellular tumor spheroid models (MCTS) are often coined as 3D in vitro models that can mimic the microenvironment of tissues. MCTS have gained increasing interest in the nano-biotechnology field as they can provide easily accessible information on the performance of nanoparticles without using animal models. Considering that many countries have put restrictions on animals testing, which will only tighten in the future as seen by the recent developments in the Netherlands, 3D models will become an even more valuable tool. Here, an overview on MCTS is provided, focusing on their use in cancer research as most nanoparticles are tested in MCTS for treatment of primary tumors. Thereafter, various types of nanoparticles-from self-assembled block copolymers to inorganic nanoparticles, are discussed. A range of physicochemical parameters including the size, shape, surface chemistry, ligands attachment, stability, and stiffness are found to influence nanoparticles in MCTS. Some of these studies are complemented by animal studies confirming that lessons from MCTS can in part predict the behaviour in vivo. In summary, MCTS are suitable models to gain additional information on nanoparticles. While not being able to replace in vivo studies, they can bridge the gap between traditional 2D in vitro studies and in vivo models.
Collapse
Affiliation(s)
- Hongxu Lu
- Centre for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Kensington, Sydney, New South Wales, 2052, Australia
| | - Martina H Stenzel
- Centre for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Kensington, Sydney, New South Wales, 2052, Australia
| |
Collapse
|
35
|
Fisusi FA, Schätzlein AG, Uchegbu IF. Nanomedicines in the treatment of brain tumors. Nanomedicine (Lond) 2018; 13:579-583. [PMID: 29376468 DOI: 10.2217/nnm-2017-0378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Funmilola A Fisusi
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.,Drug Research & Production Unit, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Andreas G Schätzlein
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.,Nanomerics Ltd., New Bridge Street House, 30-34 New Bridge Street, London, EC4V 6BJ, UK
| | - Ijeoma F Uchegbu
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.,Nanomerics Ltd., New Bridge Street House, 30-34 New Bridge Street, London, EC4V 6BJ, UK
| |
Collapse
|
36
|
Yan X, Xu L, Bi C, Duan D, Chu L, Yu X, Wu Z, Wang A, Sun K. Lactoferrin-modified rotigotine nanoparticles for enhanced nose-to-brain delivery: LESA-MS/MS-based drug biodistribution, pharmacodynamics, and neuroprotective effects. Int J Nanomedicine 2018; 13:273-281. [PMID: 29391788 PMCID: PMC5768421 DOI: 10.2147/ijn.s151475] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Efficient delivery of rotigotine into the brain is crucial for obtaining maximum therapeutic efficacy for Parkinson’s disease (PD). Therefore, in the present study, we prepared lactoferrin-modified rotigotine nanoparticles (Lf-R-NPs) and studied their biodistribution, pharmacodynamics, and neuroprotective effects following nose-to-brain delivery in the rat 6-hydroxydopamine model of PD. Materials and methods The biodistribution of rotigotine nanoparticles (R-NPs) and Lf-R-NPs after intranasal administration was assessed by liquid extraction surface analysis coupled with tandem mass spectrometry. Contralateral rotations were quantified to evaluate pharmacodynamics. Tyrosine hydroxylase and dopamine transporter immunohistochemistry were performed to compare the neuroprotective effects of levodopa, R-NPs, and Lf-R-NPs. Results Liquid extraction surface analysis coupled with tandem mass spectrometry analysis, used to examine rotigotine biodistribution, showed that Lf-R-NPs more efficiently supplied rotigotine to the brain (with a greater sustained amount of the drug delivered to this organ, and with more effective targeting to the striatum) than R-NPs. The pharmacodynamic study revealed a significant difference (P<0.05) in contralateral rotations between rats treated with Lf-R-NPs and those treated with R-NPs. Furthermore, Lf-R-NPs significantly alleviated nigrostriatal dopaminergic neurodegeneration in the rat model of 6-hydroxydopamine-induced PD. Conclusion Our findings show that Lf-R-NPs deliver rotigotine more efficiently to the brain, thereby enhancing efficacy. Therefore, Lf-R-NPs might have therapeutic potential for the treatment of PD.
Collapse
Affiliation(s)
- Xiuju Yan
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province
| | - Lixiao Xu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province
| | - Chenchen Bi
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province
| | - Dongyu Duan
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province
| | - Liuxiang Chu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province
| | - Xin Yu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province
| | - Zimei Wu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province
| | - Aiping Wang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province.,State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co., Ltd, Yantai, Shandong Province, People's Republic of China
| | - Kaoxiang Sun
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shandong Province.,State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co., Ltd, Yantai, Shandong Province, People's Republic of China
| |
Collapse
|
37
|
Li Y, Dang Y, Han D, Tan Y, Liu X, Zhang F, Xu Y, Zhang H, Yan X, Zhang X, Zeng Z, Sun X. An Angiopep-2 functionalized nanoformulation enhances brain accumulation of tanshinone IIA and exerts neuroprotective effects against ischemic stroke. NEW J CHEM 2018. [DOI: 10.1039/c8nj02441c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Effective intervention against ischemic stroke requires delivery of potent neuroprotective drugs to the brain.
Collapse
|
38
|
Li Y, An C, Han D, Dang Y, Liu X, Zhang F, Xu Y, Zhong H, Sun X. Neutrophil affinity for PGP and HAIYPRH (T7) peptide dual-ligand functionalized nanoformulation enhances the brain delivery of tanshinone IIA and exerts neuroprotective effects against ischemic stroke by inhibiting proinflammatory signaling pathways. NEW J CHEM 2018. [DOI: 10.1039/c8nj04819c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A great challenge to the therapy of ischemic stroke is the poor physicochemical properties and inability of the drug to cross the blood–brain barrier (BBB).
Collapse
Affiliation(s)
- Yutao Li
- Department of Pharmaceutical Engineering
- School of Chemical and Environmental Engineering
- Key Laboratory of Green Chemical Engineering
- Harbin University of Science and Technology
- Harbin
| | - Chiying An
- The First Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Danan Han
- Department of Pharmaceutical Engineering
- School of Chemical and Environmental Engineering
- Key Laboratory of Green Chemical Engineering
- Harbin University of Science and Technology
- Harbin
| | - Yanxin Dang
- Department of Pharmaceutical Engineering
- School of Chemical and Environmental Engineering
- Key Laboratory of Green Chemical Engineering
- Harbin University of Science and Technology
- Harbin
| | - Xin Liu
- Department of Pharmaceutical Engineering
- School of Chemical and Environmental Engineering
- Key Laboratory of Green Chemical Engineering
- Harbin University of Science and Technology
- Harbin
| | - Fengming Zhang
- Department of Pharmaceutical Engineering
- School of Chemical and Environmental Engineering
- Key Laboratory of Green Chemical Engineering
- Harbin University of Science and Technology
- Harbin
| | - Yuan Xu
- Department of Pharmacology
- School of Medicine
- Yale University
- New Haven
- USA
| | - Haijing Zhong
- Department of Pharmacology
- School of Medicine
- Yale University
- New Haven
- USA
| | - Xiaojun Sun
- Department of Pharmaceutical Engineering
- School of Chemical and Environmental Engineering
- Key Laboratory of Green Chemical Engineering
- Harbin University of Science and Technology
- Harbin
| |
Collapse
|
39
|
Kamei N, Yamaoka A, Fukuyama Y, Itokazu R, Takeda-Morishita M. Noncovalent Strategy with Cell-Penetrating Peptides to Facilitate the Brain Delivery of Insulin through the Blood–Brain Barrier. Biol Pharm Bull 2018; 41:546-554. [DOI: 10.1248/bpb.b17-00848] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Noriyasu Kamei
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| | - Ai Yamaoka
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| | - Yukiko Fukuyama
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| | - Rei Itokazu
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| | - Mariko Takeda-Morishita
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| |
Collapse
|
40
|
Yuan J, Zhou X, Cao W, Bi L, Zhang Y, Yang Q, Wang S. Improved Antitumor Efficacy and Pharmacokinetics of Bufalin via PEGylated Liposomes. NANOSCALE RESEARCH LETTERS 2017; 12:585. [PMID: 29124481 PMCID: PMC5680394 DOI: 10.1186/s11671-017-2346-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/20/2017] [Indexed: 05/24/2023]
Abstract
Bufalin was reported to show strong pharmacological effects including cardiotonic, antiviral, immune-regulation, and especially antitumor effects. The objective of this study was to determine the characterization, antitumor efficacy, and pharmacokinetics of bufalin-loaded PEGylated liposomes compared with bufalin entity, which were prepared by FDA-approved pharmaceutical excipients. Bufalin-loaded PEGylated liposomes and bufalin-loaded liposomes were prepared reproducibly with homogeneous particle size by the combination of thin film evaporation method and high-pressure homogenization method. Their mean particle sizes were 127.6 and 155.0 nm, mean zeta potentials were 2.24 and - 18.5 mV, and entrapment efficiencies were 76.31 and 78.40%, respectively. In vitro release profile revealed that the release of bufalin in bufalin-loaded PEGylated liposomes was slower than that in bufalin-loaded liposomes. The cytotoxicity of blank liposomes has been found within acceptable range, whereas bufalin-loaded PEGylated liposomes showed enhanced cytotoxicity to U251 cells compared with bufalin entity. In vivo pharmacokinetics indicated that bufalin-loaded PEGylated liposomes could extend or eliminate the half-life time of bufalin in plasma in rats. The results suggested that bufalin-loaded PEGylated liposomes improved the solubility and increased the drug concentration in plasma.
Collapse
Affiliation(s)
- Jiani Yuan
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Xuanxuan Zhou
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Wei Cao
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Linlin Bi
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yifang Zhang
- Shaanxi Pharmaceutical Development Center, Xi'an, China
| | - Qian Yang
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, The Fourth Military Medical University, Xi'an, China.
| | - Siwang Wang
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
41
|
Zhang J, Zheng Y, Xie X, Wang L, Su Z, Wang Y, Leong KW, Chen M. Cleavable Multifunctional Targeting Mixed Micelles with Sequential pH-Triggered TAT Peptide Activation for Improved Antihepatocellular Carcinoma Efficacy. Mol Pharm 2017; 14:3644-3659. [PMID: 28994600 DOI: 10.1021/acs.molpharmaceut.7b00404] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although tumor-targeting nanovehicles for hepatocellular carcinoma (HCC) chemotherapy have attracted great research and clinic interest, the poor cancer penetration, inefficient cellular uptake, and slow intracellular drug release greatly compromise their therapeutic outcomes. In this work, a multifunctional mixed micellar system, consisting of glycyrrhetinic acid (GA) for specific liver-targeting, trans-activator of transcription (TAT) peptide for potent cell penetration, and pH-sensitive poly(β-amino ester) polymers for acidic-triggered drug release, was developed to provide HCC-targeting delivery and pH-triggered release of doxorubicin (DOX). These micelles were hypothesized to efficaciously accumulate in HCC site by the guide of GA ligands, enter into cancer cells facilitated by the activated TAT peptide on the micellar surface, and finally rapidly release DOX in cytoplasm. To demonstrate this design, DOX was initially loaded in micelles modified with both GA and TAT (DOX/GA@TAT-M) with high drug loading efficiency and pH-sensitive drug release profiles. The HCC-targeting cellular uptake and synergetic anticancer efficacy were tested, indicating DOX/GA@TAT-M could be specifically and effectively internalized into HCC cells by the effect of GA targeting and TAT penetrating with enhanced cytotoxicity. In addition, the prolonged circulation time and enhanced accumulation in tumor facilitated its potent tumor growth inhibition activity in vivo. These results demonstrated that the cleavable multifunctional mixed micelles with tumor targeting, controlled TAT peptide activation, and sequential pH-sensitive drug release could be an efficient strategy for HCC treatment.
Collapse
Affiliation(s)
- Jinming Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao 999078, China
| | - Yifeng Zheng
- College of Chinese Medicines, Guangzhou University of Chinese Medicine , Guangzhou 510006, China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Sun Yat-Sen University , Guangzhou 510275, China
| | - Lan Wang
- College of Chinese Medicines, Guangzhou University of Chinese Medicine , Guangzhou 510006, China
| | - Ziren Su
- College of Chinese Medicines, Guangzhou University of Chinese Medicine , Guangzhou 510006, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao 999078, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University , New York, New York 10027, United States
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao 999078, China
| |
Collapse
|
42
|
Han W, Yin G, Pu X, Chen X, Liao X, Huang Z. Glioma targeted delivery strategy of doxorubicin-loaded liposomes by dual-ligand modification. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:1695-1712. [PMID: 28699828 DOI: 10.1080/09205063.2017.1348739] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier (BBB) is the protective parclose of brain safety, but it is also the main obstacle of the drug delivery to cerebral parenchyma, which hamper therapy for brain diseases. In this work, a glioma targeted drug delivery system was developed through loading doxorubicin into Angiopep-2 and TAT peptide dual-modified liposomes (DOX-TAT-Ang-LIP). Low-density lipoprotein receptor-related protein-1 (LRP1) was one receptor overexpressed on both BBB and glioma cytomembranes. Angiopep-2, a specific ligand of LRP1, exhibited high LRP1 binding efficiency. Additionally, TAT could penetrate through cell membranes without selectivity via an unsaturated pathway. To avoid the receptor saturation of Angiopep-2, TAT was also conjugated on the surface of liposomes, providing that the liposomes not only have effective BBB penetrating effect, but also have the glioma targeting function. The prepared DOX liposomes appeared good stability and narrow dispersity in serum with a diameter of 90 nm, and exhibited sustained DOX release behaviors. The conjunctions of Angiopep-2 and TAT were confirmed by 1H NMR spectra. The BBB model, cellular uptake observations, antiproliferation study, and the cell ultrastructure analyses suggested that DOX-TAT-Ang-LIP could not only penetrate through BBB via transcytosis, but also concentrate in glioma, then enter into glioma cells and finally result in the necrosis of glioma cells.
Collapse
Affiliation(s)
- Wei Han
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Guangfu Yin
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Ximing Pu
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Xianchun Chen
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Xiaoming Liao
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Zhongbing Huang
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| |
Collapse
|
43
|
Self-assembled amphiphilic core-shell nanocarriers in line with the modern strategies for brain delivery. J Control Release 2017. [PMID: 28648865 DOI: 10.1016/j.jconrel.2017.06.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Disorders of the central nervous system (CNS) represent increasing social and economic problems all over the world which makes the effective transport of drugs to the brain a crucial need. In the last decade, many strategies were introduced to deliver drugs to the brain trying to overcome the challenge of the blood brain barrier (BBB) using both invasive and non-invasive methods. Non-invasive strategy represented in the application of nanocarriers became very common. One of the most hopeful nanoscopic carriers for brain delivery is core-shell nanocarriers or polymeric micelles (PMs). They are more advantageous than other nanocarriers. They offer small size, ease of preparation, ease of sterilization and the possibility of surface modification with various ligands. Hence, the aim of this review is to discuss modern strategies for brain delivery, micelles as a successful delivery system for the brain and how micelles could be modified to act as "magic bullets" for brain delivery.
Collapse
|
44
|
Takechi-Haraya Y, Goda Y, Sakai-Kato K. Control of Liposomal Penetration into Three-Dimensional Multicellular Tumor Spheroids by Modulating Liposomal Membrane Rigidity. Mol Pharm 2017; 14:2158-2165. [PMID: 28410440 DOI: 10.1021/acs.molpharmaceut.7b00051] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Effective penetration of drug-carrying nanoparticles into solid tumors is a major challenge in cancer therapy. Exploration of the physicochemical properties of nanoparticles that affect penetration efficiency is required to achieve maximum therapeutic effects. Here, we used confocal laser scanning microscopy to evaluate the efficiencies of penetration of fluorescently labeled liposomes into three-dimensional spheroids composed of HeLa cells. The prepared liposomes were composed of phosphatidylcholines and varying contents of cholesterol and/or a polyethylene glycol-modified phospholipid. We demonstrated that the efficiency of penetration into spheroids increased with the bending modulus (i.e., membrane rigidity) of the liposome, as determined by atomic force microscopy (correlation coefficient, 0.84). To clarify the mechanism by which membrane rigidity contributes to the penetration behavior of liposomes, we also analyzed the cellular uptake using monolayer cells. We showed that penetration efficiency was explained partially by cellular uptake efficiency, but that other factors such as liposome diffusion efficiency in the intercellular space of tumor spheroids contributed. Our results quantitatively demonstrate that the bending modulus of the liposomal membrane is a major determinant of liposomal penetration into three-dimensional spheroids. The present study will contribute to the understanding and control of tumor penetration of liposomal formulations.
Collapse
Affiliation(s)
- Yuki Takechi-Haraya
- Division of Drugs, National Institute of Health Sciences , 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Yukihiro Goda
- Division of Drugs, National Institute of Health Sciences , 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Kumiko Sakai-Kato
- Division of Drugs, National Institute of Health Sciences , 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| |
Collapse
|
45
|
Chen C, Duan Z, Yuan Y, Li R, Pang L, Liang J, Xu X, Wang J. Peptide-22 and Cyclic RGD Functionalized Liposomes for Glioma Targeting Drug Delivery Overcoming BBB and BBTB. ACS APPLIED MATERIALS & INTERFACES 2017; 9:5864-5873. [PMID: 28128553 DOI: 10.1021/acsami.6b15831] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Chemotherapy outcomes for the treatment of glioma remain unsatisfied due to the inefficient drug transport across BBB/BBTB and poor drug accumulation in the tumor site. Nanocarriers functionalized with different targeting ligands are considered as one of the most promising alternatives. However, few studies were reported to compare the targeting efficiency of the ligands and develop nanoparticles to realize BBB/BBTB crossing and brain tumor targeting simultaneously. In this study, six peptide-based ligands (Angiopep-2, T7, Peptide-22, c(RGDfK), D-SP5 and Pep-1), widely used for brain delivery, were selected to decorate liposomes, respectively, so as to compare their targeting ability to BBB or BBTB. Based on the in vitro cellular uptake results on BCECs and HUVECs, Peptide-22 and c(RGDfK) were picked to construct a BBB/BBTB dual-crossing, glioma-targeting liposomal drug delivery system c(RGDfK)/Pep-22-DOX-LP. In vitro cellular uptake demonstrated that the synergetic effect of c(RGDfK) and Peptide-22 could significantly increase the internalization of liposomes on U87 cells. In vivo imaging further verified that c(RGDfK)/Pep-22-LP exhibited higher brain tumor distribution than single ligand modified liposomes. The median survival time of glioma-bearing mice treated with c(RGDfK)/Pep-22-DOX-LP (39.5 days) was significantly prolonged than those treated with free doxorubicin or other controls. In conclusion, the c(RGDfK) and Peptide-22 dual-modified liposome was constructed based on the targeting ability screening of various ligands. The system could effectively overcome BBB/BBTB barriers, target to tumor cells and inhibit the growth of glioma, which proved its potential for improving the efficacy of chemotherapeutics for glioma therapy.
Collapse
Affiliation(s)
- Cuitian Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Ziqing Duan
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Yan Yuan
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Ruixiang Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Liang Pang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Jianming Liang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| | - Xinchun Xu
- Shanghai Xuhui Central Hospital , Shanghai 200031, People's Republic of China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203, People's Republic of China
| |
Collapse
|
46
|
Wang Y, Qiu Y, Yin S, Zhang L, Shi K, Gao H, Zhang Z, He Q. A functional nanocarrier that copenetrates extracellular matrix and multiple layers of tumor cells for sequential and deep tumor autophagy inhibitor and chemotherapeutic delivery. Autophagy 2017; 13:359-370. [PMID: 27911136 PMCID: PMC5324845 DOI: 10.1080/15548627.2016.1256523] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/19/2016] [Accepted: 10/31/2016] [Indexed: 02/05/2023] Open
Abstract
To further enhance the intensity of deep tumor drug delivery and integrate a combined therapy, we herein report on a core-shell nanocarrier that could simultaneously overcome the double barriers of the extracellular matrix (ECM) and multiple layers of tumor cells (MLTC). A pH-triggered reversible swelling-shrinking core and an MMP2 (matrix metallopeptidase 2) degradable shell were developed to encapsulate chemotherapeutics and macroautophagy/autophagy inhibitors, respectively. MMP2 degraded the shell, which was followed by the autophagy inhibitors' release. The exposed core could diffuse along the pore within the ECM to deliver chemotherapeutics into deep tumors, and it was able to swell in lysosomes and shrink back in the cytoplasm or ECM. The swelling of the core resulted in the rapid release of chemotherapeutics to kill autophagy-inhibited cells. After leaving the dead cells, the shrinking core could act on neighboring cells that were closer to the center of the tumor. The core thus could also cross MLTC layer by layer to deliver chemotherapeutics into the deep tumor.
Collapse
Affiliation(s)
- Yang Wang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Qiu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Sheng Yin
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kairong Shi
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
Shi W, Cui X, Shi J, Chen J, Wang Y. Overcoming the blood–brain barrier for glioma-targeted therapy based on an interleukin-6 receptor-mediated micelle system. RSC Adv 2017. [DOI: 10.1039/c7ra03208k] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An interleukin-6 receptor-mediated micelle-drug system was prepared for cascade-targeting chemotherapy of glioma, which exhibited high BBB-crossing and glioma-targeted efficiency.
Collapse
Affiliation(s)
- Wei Shi
- Department of Neurosurgery
- Affiliated Hospital of Nantong University
- Nantong 226001
- China
| | - Xuexue Cui
- Center for Advanced Low-dimension Materials
- Donghua University
- Shanghai 201620
- China
| | - Jinlong Shi
- Department of Neurosurgery
- Affiliated Hospital of Nantong University
- Nantong 226001
- China
| | - Jian Chen
- Department of Neurosurgery
- Affiliated Hospital of Nantong University
- Nantong 226001
- China
| | - Yi Wang
- Center for Advanced Low-dimension Materials
- Donghua University
- Shanghai 201620
- China
- Center of Analysis and Measurement
| |
Collapse
|
48
|
Lin T, Zhao P, Jiang Y, Tang Y, Jin H, Pan Z, He H, Yang VC, Huang Y. Blood-Brain-Barrier-Penetrating Albumin Nanoparticles for Biomimetic Drug Delivery via Albumin-Binding Protein Pathways for Antiglioma Therapy. ACS NANO 2016; 10:9999-10012. [PMID: 27934069 DOI: 10.1021/acsnano.6b04268] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nutrient transporters have been explored for biomimetic delivery targeting the brain. The albumin-binding proteins (e.g., SPARC and gp60) are overexpressed in many tumors for transport of albumin as an amino acid and an energy source for fast-growing cancer cells. However, their application in brain delivery has rarely been investigated. In this work, SPARC and gp60 overexpression was found on glioma and tumor vessel endothelium; therefore, such pathways were explored for use in brain-targeting biomimetic delivery. We developed a green method for blood-brain barrier (BBB)-penetrating albumin nanoparticle synthesis, with the capacity to coencapsulate different drugs and no need for cross-linkers. The hydrophobic drugs (i.e., paclitaxel and fenretinide) yield synergistic effects to induce albumin self-assembly, forming dual drug-loaded nanoparticles. The albumin nanoparticles can penetrate the BBB and target glioma cells via the mechanisms of SPARC- and gp60-mediated biomimetic transport. Importantly, by modification with the cell-penetrating peptide LMWP, the albumin nanoparticles display enhanced BBB penetration, intratumoral infiltration, and cellular uptake. The LMWP-modified nanoparticles exhibited improved treatment outcomes in both subcutaneous and intracranial glioma models, with reduced toxic side effects. The therapeutic mechanisms were associated with induction of apoptosis, antiangiogenesis, and tumor immune microenvironment regulation. It provides a facile method for dual drug-loaded albumin nanoparticle preparation and a promising avenue for biomimetic delivery targeting the brain tumor based on combination therapy.
Collapse
Affiliation(s)
- Tingting Lin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University , Tianjin 300070, China
- Department of Pharmacy, Binzhou Medical University Hospital , 661 Huanghe Road, Binzhou 256603, China
| | - Pengfei Zhao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
- Nanchang University College of Pharmacy , 461 Bayi Road, Nanchang 330006, China
| | - Yifan Jiang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Yisi Tang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Hongyue Jin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Zhenzhen Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University , Tianjin 300070, China
| | - Victor C Yang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University , Tianjin 300070, China
- University of Michigan College of Pharmacy , 428 Church Street, Ann Arbor, Michigan 48108, United States
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| |
Collapse
|
49
|
Bi Y, Liu L, Lu Y, Sun T, Shen C, Chen X, Chen Q, An S, He X, Ruan C, Wu Y, Zhang Y, Guo Q, Zheng Z, Liu Y, Lou M, Zhao S, Jiang C. T7 Peptide-Functionalized PEG-PLGA Micelles Loaded with Carmustine for Targeting Therapy of Glioma. ACS APPLIED MATERIALS & INTERFACES 2016; 8:27465-27473. [PMID: 27466824 DOI: 10.1021/acsami.6b05572] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Glioma is regarded as the deadliest and most common brain tumor because of the extremely difficult surgical excision ascribed from its invasive nature. In addition, the natural blood-brain barrier (BBB) greatly restricts the therapeutics' penetration into the central nervous system. Carmustine (BCNU) is a widely used antiglioma drug in clinical applications. However, its serious complications prevent it from being applied in a clinical setting to some extent. Thus, it is urgent to explore novel BCNU delivery systems specially designed for glioma. Development of polymeric nanoparticles offers a favorable alternative to serve this purpose. Particularly, use of poly(lactic-co-glycolic acid) (PLGA) has been shown to be advantageous for its favorable biodegradability and biocompatibility, which ensure safe therapies. In this study, T7 peptide-conjugated, BCNU-loaded micelles were constructed successfully via the emulsion-solvent evaporation method. The micelles were characterized by transmission electron microscopy and dynamic light scattering in detail, and the capacity of BBB crossing was studied. The in vivo detecting results of the targeting effect using the BODIPY probe evidenced that T7-modified micelles showed a more pronounced accumulation and accumulated in the tumor more efficiently than in the unconjugated probe. Meanwhile, the targeting group exhibited the best curative effect accompanied with the lowest loss in body weight, the smallest tumor size, and an obviously prolonged survival time among the groups. In the near future, we believe the targeted delivery system specially designed for BCNU is expected to provide sufficient evidence to proceed to clinical trials.
Collapse
Affiliation(s)
- Yunke Bi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University , Harbin, Heilongjiang 150001, P.R. China
- Department of Neurosurgery, Shanghai First People's Hospital, Shanghai Jiao Tong University, School of Medicine , Shanghai 201620, P.R. China
| | - Lisha Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Yifei Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Chen Shen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University , Harbin, Heilongjiang 150001, P.R. China
| | - Xinli Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Qinjun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Sai An
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Xi He
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Chunhui Ruan
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Yinhao Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Yujie Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Qin Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Zhixing Zheng
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University , Harbin, Heilongjiang 150001, P.R. China
| | - Yaohua Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University , Harbin, Heilongjiang 150001, P.R. China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai First People's Hospital, Shanghai Jiao Tong University, School of Medicine , Shanghai 201620, P.R. China
| | - Shiguang Zhao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University , Harbin, Heilongjiang 150001, P.R. China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, P.R. China
| |
Collapse
|
50
|
Vieira DB, Gamarra LF. Getting into the brain: liposome-based strategies for effective drug delivery across the blood-brain barrier. Int J Nanomedicine 2016; 11:5381-5414. [PMID: 27799765 PMCID: PMC5077137 DOI: 10.2147/ijn.s117210] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review summarizes articles that have been reported in literature on liposome-based strategies for effective drug delivery across the blood–brain barrier. Due to their unique physicochemical characteristics, liposomes have been widely investigated for their application in drug delivery and in vivo bioimaging for the treatment and/or diagnosis of neurological diseases, such as Alzheimer’s, Parkinson’s, stroke, and glioma. Several strategies have been used to deliver drug and/or imaging agents to the brain. Covalent ligation of such macromolecules as peptides, antibodies, and RNA aptamers is an effective method for receptor-targeting liposomes, which allows their blood–brain barrier penetration and/or the delivery of their therapeutic molecule specifically to the disease site. Additionally, methods have been employed for the development of liposomes that can respond to external stimuli. It can be concluded that the development of liposomes for brain delivery is still in its infancy, although these systems have the potential to revolutionize the ways in which medicine is administered.
Collapse
Affiliation(s)
| | - Lionel F Gamarra
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| |
Collapse
|