1
|
Llaguno-Munive M, Vazquez-Lopez MI, Garcia-Lopez P. Solid Lipid Nanoparticles, an Alternative for the Treatment of Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:10712. [PMID: 39409041 PMCID: PMC11476567 DOI: 10.3390/ijms251910712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Within the field of nanomedicine, which is revolutionizing cancer treatment, solid lipid nanoparticles (SLNs) have shown advantages over conventional chemotherapy when tested on cancer cells in preclinical studies. SLNs have proven to be an innovative strategy for the treatment of triple-negative breast cancer cells, providing greater efficiency than existing treatments in various studies. The encapsulation of antineoplastic drugs in SLNs has facilitated a sustained, controlled, and targeted release, which enhances therapeutic efficiency and reduces adverse effects. Moreover, the surface of SLNs can be modified to increase efficiency. For instance, the coating of these particles with polyethylene glycol (PEG) decreases their opsonization, resulting in a longer life in the circulatory system. The creation of positively charged cationic SLNs (cSLNs), achieved by the utilization of surfactants or ionic lipids with positively charged structural groups, increases their affinity for cell membranes and plasma proteins. Hyaluronic acid has been added to SLNs so that the distinct pH of tumor cells would stimulate the release of the drug and/or genetic material. The current review summarizes the recent research on SLNs, focusing on the encapsulation and transport of therapeutic agents with a cytotoxic effect on triple-negative breast cancer.
Collapse
Affiliation(s)
- Monserrat Llaguno-Munive
- Laboratorio de Física Médica, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| | - Maria Ines Vazquez-Lopez
- Laboratorio de Fármaco-Oncología y Nanomedicina, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| | - Patricia Garcia-Lopez
- Laboratorio de Fármaco-Oncología y Nanomedicina, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| |
Collapse
|
2
|
Attri N, Das S, Banerjee J, Shamsuddin SH, Dash SK, Pramanik A. Liposomes to Cubosomes: The Evolution of Lipidic Nanocarriers and Their Cutting-Edge Biomedical Applications. ACS APPLIED BIO MATERIALS 2024; 7:2677-2694. [PMID: 38613498 PMCID: PMC11110070 DOI: 10.1021/acsabm.4c00153] [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: 02/01/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024]
Abstract
Lipidic nanoparticles have undergone extensive research toward the exploration of their diverse therapeutic applications. Although several liposomal formulations are in the clinic (e.g., DOXIL) for cancer therapy, there are many challenges associated with traditional liposomes. To address these issues, modifications in liposomal structure and further functionalization are desirable, leading to the emergence of solid lipid nanoparticles and the more recent liquid lipid nanoparticles. In this context, "cubosomes", third-generation lipidic nanocarriers, have attracted significant attention due to their numerous advantages, including their porous structure, structural adaptability, high encapsulation efficiency resulting from their extensive internal surface area, enhanced stability, and biocompatibility. Cubosomes offer the potential for both enhanced cellular uptake and controlled release of encapsulated payloads. Beyond cancer therapy, cubosomes have demonstrated effectiveness in wound healing, antibacterial treatments, and various dermatological applications. In this review, the authors provide an overview of the evolution of lipidic nanocarriers, spanning from conventional liposomes to solid lipid nanoparticles, with a special emphasis on the development and application of cubosomes. Additionally, it delves into recent applications and preclinical trials associated with cubosome formulations, which could be of significant interest to readers from backgrounds in nanomedicine and clinicians.
Collapse
Affiliation(s)
- Nishtha Attri
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | - Swarnali Das
- Department
of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | - Jhimli Banerjee
- Department
of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | - Shazana H. Shamsuddin
- Department
of Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Sandeep Kumar Dash
- Department
of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | - Arindam Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
- School
of Medicine, University of Leeds, Leeds LS53RL, United Kingdom
| |
Collapse
|
3
|
Li J, Zhang H, Mao X, Deng H, Fan L, Yue L, Li C, Pan S, Wen X. Preparation, in vitro anti-tumour activity and in vivo pharmacokinetics of RGD-decorated liposomes loaded with shikonin. Pharm Dev Technol 2024; 29:153-163. [PMID: 38330994 DOI: 10.1080/10837450.2024.2315457] [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: 08/17/2023] [Accepted: 02/03/2024] [Indexed: 02/10/2024]
Abstract
Shikonin (SHK) has been evidenced to possess effects against various cancer cells. However, poor aqueous solubility and high toxicity restrict its application. In the study, RGD-decorated liposomes loaded with SHK (RGD-Lipo-SHK) were prepared via thin-film hydration method. Characterization and cellular uptake of liposomes was evaluated. Cytotoxicity of blank liposomes and different SHK formulations was measured against breast cancer cells (MDA-MB-231, MCF-7, and MCF-10A). Anti-tumour effects and pharmacokinetic parameters of different SHK formulations were appraised in tumour spheroids and in rat model, respectively. Liposomes displayed a particle size of less than 127 nm with a polydispersity index about 0.21. The encapsulation efficiency was about 91% for SHK, and drug leakage rate of liposomes was less than 6%. RGD-Lipo-SHK showed superior cellular internalization in the αvβ3-positive MDA-MB-231 cells. Blank liposomes had no cytotoxicity to MDA-MB-231 and MCF-7 cells. Howbeit, different SHK formulations obviously inhibited proliferation of MCF-10A cells, especially free SHK. Meanwhile, RGD-Lipo-SHK significantly inhibited growth inhibition of tumour spheroids. The pharmacokinetics study indicated that the peak concentration, area under plasma concentration-time curves, half-life, and mean residence time of RGD-Lipo-SHK distinctly increased compared with those of free SHK. Altogether, these results demonstrated RGD-Lipo-SHK could reduce cytotoxicity, strengthen the antitumor-targeted effect, and prolong circulation time, which provides a foundation for further in vivo experimentations.
Collapse
Affiliation(s)
- Jiping Li
- Public Health School, Qiqihar Medical University, Qiqihar, China
| | - Hao Zhang
- Pharmacy School, Qiqihar Medical University, Qiqihar, China
| | - Xinliang Mao
- Pharmacy School, Qiqihar Medical University, Qiqihar, China
| | - Huilin Deng
- Pharmacy School, Qiqihar Medical University, Qiqihar, China
| | - Li Fan
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Liling Yue
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Chengchong Li
- Mental Health School, Qiqihar Medical University, Qiqihar, China
| | - Siwen Pan
- Pathology School, Qiqihar Medical University, Qiqihar, China
| | - Xianchun Wen
- Medical Techinology School, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
4
|
Huang M, Zhai BT, Fan Y, Sun J, Shi YJ, Zhang XF, Zou JB, Wang JW, Guo DY. Targeted Drug Delivery Systems for Curcumin in Breast Cancer Therapy. Int J Nanomedicine 2023; 18:4275-4311. [PMID: 37534056 PMCID: PMC10392909 DOI: 10.2147/ijn.s410688] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/19/2023] [Indexed: 08/04/2023] Open
Abstract
Breast cancer (BC) is the most prevalent type of cancer in the world and the main reason women die from cancer. Due to the significant side effects of conventional treatments such as chemotherapy and radiotherapy, the search for supplemental and alternative natural drugs with lower toxicity and side effects is of interest to researchers. Curcumin (CUR) is a natural polyphenol extracted from turmeric. Numerous studies have demonstrated that CUR is an effective anticancer drug that works by modifying different intracellular signaling pathways. CUR's therapeutic utility is severely constrained by its short half-life in vivo, low water solubility, poor stability, quick metabolism, low oral bioavailability, and potential for gastrointestinal discomfort with high oral doses. One of the most practical solutions to the aforementioned issues is the development of targeted drug delivery systems (TDDSs) based on nanomaterials. To improve drug targeting and efficacy and to serve as a reference for the development and use of CUR TDDSs in the clinical setting, this review describes the physicochemical properties and bioavailability of CUR and its mechanism of action on BC, with emphasis on recent studies on TDDSs for BC in combination with CUR, including passive TDDSs, active TDDSs and physicochemical TDDSs.
Collapse
Affiliation(s)
- Mian Huang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Bing-Tao Zhai
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Yu Fan
- School of Basic Medicine, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jing Sun
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Ya-Jun Shi
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Xiao-Fei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jun-Bo Zou
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jia-Wen Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Dong-Yan Guo
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| |
Collapse
|
5
|
Sanati M, Afshari AR, Aminyavari S, Kesharwani P, Jamialahmadi T, Sahebkar A. RGD-engineered nanoparticles as an innovative drug delivery system in cancer therapy. J Drug Deliv Sci Technol 2023; 84:104562. [DOI: 10.1016/j.jddst.2023.104562] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
|
6
|
Lopes C, Cristóvão J, Silvério V, Lino PR, Fonte P. Microfluidic production of mRNA-loaded lipid nanoparticles for vaccine applications. Expert Opin Drug Deliv 2022; 19:1381-1395. [PMID: 36223174 DOI: 10.1080/17425247.2022.2135502] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION During past years, lipid nanoparticles (LNPs) have emerged as promising carriers for RNA delivery, with several clinical trials focusing on both infectious diseases and cancer. More recently, the success of messenger RNA (mRNA) vaccines for the treatment of severe diseases such as acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is partially justified by the development of LNPs encapsulating mRNA for efficient cytosolic delivery. AREAS COVERED This review examines the production and formulation of LNPs by using microfluidic devices, the status of mRNA-loaded LNPs therapeutics and explores spray drying process, as a promising dehydration process to enhance LNP stability and provide alternative administration routes. EXPERT OPINION Microfluidic techniques for preparation of LNPs based on organic solvent injection method promotes the generation of stable, uniform, and monodispersed nanoparticles enabling higher encapsulation efficiency. In particular, the application of microfluidics for the fabrication of mRNA-loaded LNPs is based on rapid mixing of small volumes of ethanol solution containing lipids and aqueous solution containing mRNA. Control of operating parameters and formulation has enabled the optimization of nanoparticle physicochemical characteristics and encapsulation efficiency.
Collapse
Affiliation(s)
- Carolina Lopes
- iBB - Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,Hovione Farmaciência S.A., R&D Analytical Development, Lumiar Campus, Building R,1649-038 Lisbon, Portugal.,Hovione Farmaciência S.A., R&D Inhalation and Advance Drug Delivery, Lumiar Campus, Building R, 1649-038 Lisbon, Portugal
| | - Joana Cristóvão
- Hovione Farmaciência S.A., R&D Inhalation and Advance Drug Delivery, Lumiar Campus, Building R, 1649-038 Lisbon, Portugal
| | - Vânia Silvério
- Institute of Systems and Computer Engineering for Microsystems and Nanotechnologies, INESC MN, 1000-029 Lisbon, Portugal.,Department of Physics, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal
| | - Paulo Roque Lino
- Hovione Farmaciência S.A., R&D Inhalation and Advance Drug Delivery, Lumiar Campus, Building R, 1649-038 Lisbon, Portugal
| | - Pedro Fonte
- iBB - Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,Center of Marine Sciences (CCMAR), University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal.,Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal
| |
Collapse
|
7
|
Pollalis D, Kim D, Nair GKG, Kang C, Nanda AV, Lee SY. Intraocular RGD-Engineered Exosomes and Active Targeting of Choroidal Neovascularization (CNV). Cells 2022; 11:cells11162573. [PMID: 36010651 PMCID: PMC9406786 DOI: 10.3390/cells11162573] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose: To assess the transretinal penetration of intravitreally injected retinal multicell-derived exosomes and to develop exosome-based active targeting of choroidal neovascularization (CNV) by bioengineering with ASL, which is composed of a membrane Anchor (BODIPY), Spacer (PEG), and targeting Ligands (cyclic RGD peptide). Methods: Retinal multicell-derived exosomes were recovered from a whole mouse retina using differential ultracentrifugation. Their size, number, and morphology were characterized using nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM). Exosome markers were confirmed using an exosome detection antibody array. Intravitreal injection of fluorescent (PKH-26)-labeled or engineered ASL exosomes (1 × 106 exosomes/μL) were given to the wild-type mouse or laser-induced CNV mouse model. Retinal uptake of exosomes was assessed by in vivo retinal imaging microscopy and histological staining with DAPI, GSA, and anti-integrin αv for retinal sections or choroid/RPE flat mounts. Active targeting of CNV was assessed by comparing retinal uptake between areas with and without CNV and by colocalization analysis of ASL exosomes with integrin αv within CNV. Staining with anti-F4/80, anti-ICAM-1, and anti-GFAP antibodies on retinal sections were performed to identify intracellular uptake of exosomes and immediate reactive retinal gliosis after exosome treatment. Results: An average of 2.1 × 109 particles/mL with a peak size of 140 nm exosomes were recovered. Rapid retinal penetration of intravitreally injected exosomes was confirmed by retinal imaging microscopy at 3 and 24 h post-injection. Intravitreally delivered PKH-26-labeled exosomes reached inner and outer retinal layers including IPL, INL, OPL, and ONL at 1 and 7 days post-injection. Intravitreally injected ASL exosomes were predominantly delivered to the area of CNV including ONL, RPE, and choroid in laser-induced CNV mouse models with 89.5% of colocalization with integrin αv. Part of exosomes was also taken intracellularly to vascular endothelial cells and macrophages. After intravitreal injection, neither naive exosomes nor ASL exosomes induced immediate reactive gliosis. Conclusions: Intravitreally delivered retinal multicell-derived exosomes have good retinal penetration, and ASL modification of exosomes actively targets CNV with no immediate reactive gliosis. ASL exosomes have a great potential to serve as an intraocular drug delivery vehicle, allowing an active targeting strategy.
Collapse
Affiliation(s)
- Dimitrios Pollalis
- USC Roski Eye Institute, USC Ginsburg Institute for Biomedical Therapeutics, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Dongin Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Gopa Kumar Gopinadhan Nair
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Changsun Kang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Arjun V. Nanda
- College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Sun Young Lee
- USC Roski Eye Institute, USC Ginsburg Institute for Biomedical Therapeutics, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
8
|
Dhilip Kumar SS, Abrahamse H. Recent advances in the development of biocompatible nanocarriers and their cancer cell targeting efficiency in photodynamic therapy. Front Chem 2022; 10:969809. [PMID: 36046728 PMCID: PMC9420852 DOI: 10.3389/fchem.2022.969809] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, the role of biocompatible nanocarriers (BNs) and their cancer cell targeting efficiency in photodynamic therapy (PDT) holds potential benefits for cancer treatment. Biocompatible and biodegradable nanoparticles are successfully used as carrier molecules to deliver cancer drugs and photosensitizers due to their material safety in the drug delivery system. Biocompatible nanocarriers are non-toxic and ensure high-level biocompatibility with blood, cells, and physiological conditions. The physicochemical properties of BNs often enable them to modify their surface chemistry, which makes conjugating specific ligands or antibodies to achieve cancer cell targeting drug delivery in PDT. This review article focuses on the various types of BNs used in targeted drug delivery, physicochemical properties, and surface chemistry of BNs in targeted drug delivery, advantages of BNs in drug delivery systems, and the targeting efficiency of BNs on some specific targeting receptors for cancer therapy. Furthermore, the review briefly recaps the nanocarrier-based targeted approaches in cancer PDT.
Collapse
|
9
|
Malik Z, Parveen R, Abass S, Irfan Dar M, Husain SA, Ahmad S. Receptor-Mediated Targeting in Breast Cancer through Solid Lipid Nanoparticles and Its Mechanism. Curr Drug Metab 2022; 23:800-817. [PMID: 35430962 DOI: 10.2174/1389200223666220416213639] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/20/2022] [Accepted: 03/01/2022] [Indexed: 01/05/2023]
Abstract
Nanoparticles have gained prominence in many areas and domains worldwide, such as metallic NP, carbon dots, quantum dots, polymeric NP, nano-suspension, nanocrystals, solid lipid nanoparticles (SLN), etc. and have been applied in the field of medicine as nanomedicine with promising results. Rise in cancer mortality rate has been an issue for a long time with female breast cancer as one of the most detected cancers. No permanent treatment has been developed till date could combat breast cancer with minimum side effects that are not long-lasting as there is no proper technique through which the anticancer drugs can recognize benign or malignant or normal cells that causes systematic toxicity. Advancement in technology has led to the discovery of many biological pathways and mechanisms. Tumor cells or cancer cells overexpress some high-affinity receptors that can be targeted to deliver the anticancer drugs at specific site using these pathways and mechanisms. Solid lipid nanoparticles (SLN) are among some of the excellent drug delivery systems, especially stealth SLN (sSLN). SLN, when conjugated with a ligand (called as sSLN), has affinity and specificity towards a specific receptor, and can deliver the drug in breast cancer cells overexpressing the receptors. Using this technique, various investigations have reported better anti-breast cancer activity than simple SLN (non-conjugated to ligand or no receptor targeting). This review includes the investigations and data on receptor-mediated targeting in breast cancer from 2010 to 2021 by searching different databases. Overall, information on SLN in different cancers is reviewed. In vivo investigations, pharmacokinetics, biodistribution, and stability are discussed to describe the efficacy of sSLN. Investigations included in this review demonstrate that sSLN delivers the drug by overcoming the biological barriers and shows enhanced and better activity than non-conjugated SLN which also verifies that a lesser concentration of drug can show anti-breast cancer activity. The efficacy of medicines could be increased with lower cancer deaths through stealth-SLN. Due to the low cost of synthesis, biocompatibility and easy to formulate, more study is needed in vitro and in vivo so that this novel technique could be utilized in the treatment of human breast cancer.
Collapse
Affiliation(s)
- Zoya Malik
- Human Genetics Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi-110025, India.,Bioactive Natural Product Laboratory, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Rabea Parveen
- Human Genetics Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi-110025, India.,Bioactive Natural Product Laboratory, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Sageer Abass
- Human Genetics Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi-110025, India.,Bioactive Natural Product Laboratory, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Mohammad Irfan Dar
- Bioactive Natural Product Laboratory, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India.,Proteomics and Bioinformatics Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi-110025, India
| | - Syed Akhtar Husain
- Human Genetics Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi-110025, India
| | - Sayeed Ahmad
- Bioactive Natural Product Laboratory, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| |
Collapse
|
10
|
Functionalized chitosan as a promising platform for cancer immunotherapy: A review. Carbohydr Polym 2022; 290:119452. [DOI: 10.1016/j.carbpol.2022.119452] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/20/2022]
|
11
|
Ahmed T, Liu FCF, Lu B, Lip H, Park E, Alradwan I, Liu JF, He C, Zetrini A, Zhang T, Ghavaminejad A, Rauth AM, Henderson JT, Wu XY. Advances in Nanomedicine Design: Multidisciplinary Strategies for Unmet Medical Needs. Mol Pharm 2022; 19:1722-1765. [PMID: 35587783 DOI: 10.1021/acs.molpharmaceut.2c00038] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Globally, a rising burden of complex diseases takes a heavy toll on human lives and poses substantial clinical and economic challenges. This review covers nanomedicine and nanotechnology-enabled advanced drug delivery systems (DDS) designed to address various unmet medical needs. Key nanomedicine and DDSs, currently employed in the clinic to tackle some of these diseases, are discussed focusing on their versatility in diagnostics, anticancer therapy, and diabetes management. First-hand experiences from our own laboratory and the work of others are presented to provide insights into strategies to design and optimize nanomedicine- and nanotechnology-enabled DDS for enhancing therapeutic outcomes. Computational analysis is also briefly reviewed as a technology for rational design of controlled release DDS. Further explorations of DDS have illuminated the interplay of physiological barriers and their impact on DDS. It is demonstrated how such delivery systems can overcome these barriers for enhanced therapeutic efficacy and how new perspectives of next-generation DDS can be applied clinically.
Collapse
Affiliation(s)
- Taksim Ahmed
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Fuh-Ching Franky Liu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Brian Lu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - HoYin Lip
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Elliya Park
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Ibrahim Alradwan
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Jackie Fule Liu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Chunsheng He
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Abdulmottaleb Zetrini
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Tian Zhang
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Amin Ghavaminejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Andrew M Rauth
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Jeffrey T Henderson
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
12
|
Picheth GF, Ganzella FADO, Filizzola JO, Canquerino YK, Cardoso GC, Collini MB, Colauto LB, Figueroa-Magalhães MC, Cavalieri EA, Klassen G. Ligand-mediated nanomedicines against breast cancer: a review. Nanomedicine (Lond) 2022; 17:645-664. [PMID: 35438008 DOI: 10.2217/nnm-2021-0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ligand-mediated targeting represents the cutting edge in precision-guided therapy for several diseases. Surface engineering of nanomedicines with ligands exhibiting selective or tailored affinity for overexpressed biomolecules of a specific disease may increase therapeutic efficiency and reduce side effects and recurrence. This review focuses on newly developed approaches and strategies to improve treatment and overcome the mechanisms associated with breast cancer resistance.
Collapse
Affiliation(s)
- Guilherme F Picheth
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil.,School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Paraná, Brazil
| | | | - João Oc Filizzola
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Yan K Canquerino
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Gabriela C Cardoso
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Michelle B Collini
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Leonardo B Colauto
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | | | - Edneia Asr Cavalieri
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Giseli Klassen
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| |
Collapse
|
13
|
Yang L, Tang J, Yin H, Yang J, Xu B, Liu Y, Hu Z, Yu B, Xia F, Zou G. Self-Assembled Nanoparticles for Tumor-Triggered Targeting Dual-Mode NIRF/MR Imaging and Photodynamic Therapy Applications. ACS Biomater Sci Eng 2022; 8:880-892. [PMID: 35099181 DOI: 10.1021/acsbiomaterials.1c01418] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In this study, the self-assembling strategy was used to synthesize a therapeutic and diagnostic nanosystem for tumor-triggered targeting dual-mode near-infrared fluorescence (NIRF)/magnetic resonance (MR) imaging and photodynamic therapy applications. This theranostic nanosystem was synthesized based on the self-assembling of the short peptide (PLGVRGRGDC) and the gadolinium chelator (diethylenetriamine pentaacetic acid) functionalized amphiphilic DSPE-PEG2000, followed by loading with the insoluble photosensitizer therapeutic agent chlorin e6 (Ce6). The formed theranostic nanosystem can accumulate in the matrix metalloproteinase 2 (MMP2) rich tumor sites guided by the enhanced permeability and retention effect and MMP2-substrate peptide (PLGVR) targeting. After PLGVR was hydrolyzed in the tumor microenvironment by MMP2, the nanosystem was actively taken up by tumor cells via Arg-Gly-Asp (RGD) peptide-mediated internalization. With the coexistence of gadolinium and Ce6, the formed nanosystem can be used for both NIRF/MR dual-mode imaging and photodynamic therapy. These tumor-triggered targeting self-assembled nanoparticles with low cytotoxicity and high endocytosis efficiency can efficiently induce A549 cancer cell apoptosis under laser irradiation. Meanwhile, they possessed enhanced tumor-targeted NIRF/MR imaging ability and efficiently inhibited tumor growth with minimal side effects in mice bearing A549 lung cancer. Therefore, these self-assembled theranostic nanoparticles may have great potential for cancer clinical diagnosis and therapy.
Collapse
Affiliation(s)
- Lun Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi, 330006, China
| | - Jian Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi, 330006, China
| | - Hui Yin
- Medical Department of Nanchang University, Nanchang 330006, China
| | - Jie Yang
- Medical Department of Nanchang University, Nanchang 330006, China
| | - Bin Xu
- Medical Department of Nanchang University, Nanchang 330006, China
| | - Yunkun Liu
- Medical Department of Nanchang University, Nanchang 330006, China
| | - Zhi Hu
- Medical Department of Nanchang University, Nanchang 330006, China
| | - Bentong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi, 330006, China
| | - Fangfang Xia
- Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China
| | - Guowen Zou
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
14
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
15
|
Seyyednia E, Oroojalian F, Baradaran B, Mojarrad JS, Mokhtarzadeh A, Valizadeh H. Nanoparticles modified with vasculature-homing peptides for targeted cancer therapy and angiogenesis imaging. J Control Release 2021; 338:367-393. [PMID: 34461174 DOI: 10.1016/j.jconrel.2021.08.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 10/20/2022]
Abstract
The two major challenges in cancer treatment include lack of early detection and ineffective therapies with various side effects. Angiogenesis is the key process in the growth, survival, invasiveness, and metastasis of many of cancerous tumors. Imaging of the angiogenesis could lead to diagnosis of tumors in the early stage and evaluation of the therapeutic responses. Angiogenic blood vessels express specific molecular markers different from normal blood vessels (in level or kind). This fact would make the tumor vasculature a suitable site to target therapeutics and imaging agents within the tumor. Surface modified nanoparticles using peptide ligands with high binding affinity to the vasculature markers, provide efficient delivery of therapeutic and imaging agents, while avoiding undesirable side effects. In this review, we discuss discoveries of various tumor targeting peptides useful for tumor angiogenesis imaging and targeted therapy with emphasis on surface modified nanomedicines using vasculature targeting peptides.
Collapse
Affiliation(s)
- Elham Seyyednia
- Student Research Committee and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Shahbazi Mojarrad
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hadi Valizadeh
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Slack RJ, Macdonald SJF, Roper JA, Jenkins RG, Hatley RJD. Emerging therapeutic opportunities for integrin inhibitors. Nat Rev Drug Discov 2021; 21:60-78. [PMID: 34535788 PMCID: PMC8446727 DOI: 10.1038/s41573-021-00284-4] [Citation(s) in RCA: 210] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Integrins are cell adhesion and signalling proteins crucial to a wide range of biological functions. Effective marketed treatments have successfully targeted integrins αIIbβ3, α4β7/α4β1 and αLβ2 for cardiovascular diseases, inflammatory bowel disease/multiple sclerosis and dry eye disease, respectively. Yet, clinical development of others, notably within the RGD-binding subfamily of αv integrins, including αvβ3, have faced significant challenges in the fields of cancer, ophthalmology and osteoporosis. New inhibitors of the related integrins αvβ6 and αvβ1 have recently come to the fore and are being investigated clinically for the treatment of fibrotic diseases, including idiopathic pulmonary fibrosis and nonalcoholic steatohepatitis. The design of integrin drugs may now be at a turning point, with opportunities to learn from previous clinical trials, to explore new modalities and to incorporate new findings in pharmacological and structural biology. This Review intertwines research from biological, clinical and medicinal chemistry disciplines to discuss historical and current RGD-binding integrin drug discovery, with an emphasis on small-molecule inhibitors of the αv integrins. Integrins are key signalling molecules that are present on the surface of subsets of cells and are therefore good potential therapeutic targets. In this Review, Hatley and colleagues discuss the development of integrin inhibitors, particularly the challenges in developing inhibitors for integrins that contain an αv-subunit, and suggest how these challenges could be addressed.
Collapse
Affiliation(s)
| | | | | | - R G Jenkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
17
|
Khabazian E, Vakhshiteh F, Norouzi P, Fatahi Y, Dinarvand R, Atyabi F. Cationic Liposome Decorated with Cyclic RGD Peptide for Targeted Delivery of anti-STAT3 siRNA to Melanoma Cancer Cells. J Drug Target 2021; 30:522-533. [PMID: 34482780 DOI: 10.1080/1061186x.2021.1973481] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Gene therapy is regarded as a valuable strategy for efficient cancer treatment. However, the design of effective delivery systems that can deliver gene materials such as siRNA specifically to the tumour tissues plays a pivotal role in cancer therapy. For this reason, a targeted cationic liposome for melanoma treatment was developed. This system consists of cyclic RGD peptide conjugated to DSPE-PEG2000, cholesterol, DOTAP, and DSPC as cationic and neutral lipids, respectively. Cyclic RGD was selected based on speculation that cyclic RGD would effectively transport anti-signal transducer and activator of transcription 3 (STAT3) siRNA into melanoma cell via integrin receptors. The prepared liposomes provided excellent stability against electrolyte and serum nucleases. Targeted liposomes remarkably exhibited higher cellular internalisation in comparison with the non-targeted system in flow cytometry and confocal microscopy. Furthermore, incorporating peptide on the surface of liposomes resulted in considerably high cytotoxicity, a 2.1-times raise in apoptosis induction, and a significantly enhanced STAT3 gene suppression as compared with the corresponding non-targeted formulation on B16F10 murine melanoma cells. Whole-body imaging confirmed the more significant tumour accumulation of targeted liposomes in B16F10 melanoma xenograft tumour-bearing mice. Consequently, c-RGD peptide modified liposome suggests a promising option for specific siRNA delivery into melanoma cells.
Collapse
Affiliation(s)
- Ehsan Khabazian
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 1417614411, Tehran, Iran
| | - Faezeh Vakhshiteh
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Norouzi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 1417614411, Tehran, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 1417614411, Tehran, Iran.,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 1417614411, Tehran, Iran.,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Zou T, Lu W, Mezhuev Y, Lan M, Li L, Liu F, Cai T, Wu X, Cai Y. A review of nanoparticle drug delivery systems responsive to endogenous breast cancer microenvironment. Eur J Pharm Biopharm 2021; 166:30-43. [PMID: 34098073 DOI: 10.1016/j.ejpb.2021.05.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/24/2021] [Accepted: 05/31/2021] [Indexed: 12/26/2022]
Abstract
Breast cancer, as a malignant disease that seriously threatens women's health, urgently needs to be researched to develop effective and safe therapeutic drugs. Nanoparticle drug delivery systems (NDDS), provide a powerful means for drug targeting to the breast cancer, enhancing the bioavailability and reducing the adverse effects of anticancer drug. However, the breast cancer microenvironment together with heterogeneity of cancer, impedes the tumor targeting effect of NDDS. Breast cancer microenvironment, exerts endogenous stimuli, such as hypoxia, acidosis, and aberrant protease expression, shape a natural shelter for tumor growth, invasion and migration. On the basis of the ubiquitous of endogenous stimuli in the breast cancer microenvironment, researchers exploited them to design the stimuli-responsive NDDS, which response to endogenous stimulus, targeted release drug in breast cancer microenvironment. In this review, we highlighted the effect of the breast cancer microenvironment, summarized innovative NDDS responsive to the internal stimuli in the tumor microenvironment, including the material, the targeting groups, the loading drugs, targeting position and the function of stimuli-responsive nanoparticle drug delivery system. The limitations and potential applications of the stimuli-responsive nanoparticle drug delivery systems for breast cancer treatment were discussed to further the application.
Collapse
Affiliation(s)
- Tengteng Zou
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Wenping Lu
- Guang an'men Hospital China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yaroslav Mezhuev
- Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russia
| | - Meng Lan
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Lihong Li
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Fengjie Liu
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Tiange Cai
- College of Life Sciences, Liaoning University, Shenyang 110036, PR China.
| | - Xiaoyu Wu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada.
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China; Guangdong Key Lab of Traditional Chinese Medicine Information Technology, Jinan University, Guangzhou 510632, PR China; Cancer Research Institute, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
19
|
Rahman S, Kumar V, Kumar A, Abdullah TS, Rather IA, Jan AT. Molecular Perspective of Nanoparticle Mediated Therapeutic Targeting in Breast Cancer: An Odyssey of Endoplasmic Reticulum Unfolded Protein Response (UPR ER) and Beyond. Biomedicines 2021; 9:biomedicines9060635. [PMID: 34199484 PMCID: PMC8229605 DOI: 10.3390/biomedicines9060635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is the second most frequent cause of death among women. Representing a complex and heterogeneous type of cancer, its occurrence is attributed by both genetic (gene mutations, e.g., BRCA1, BRCA2) and non-genetic (race, ethnicity, etc.) risk factors. The effectiveness of available treatment regimens (small molecules, cytotoxic agents, and inhibitors) decreased due to their poor penetration across biological barriers, limited targeting, and rapid body clearance along with their effect on normal resident cells of bone marrow, gastrointestinal tract, and hair follicles. This significantly reduced their clinical outcomes, which led to an unprecedented increase in the number of cases worldwide. Nanomedicine, a nano-formulation of therapeutics, emerged as a versatile delivering module for employment in achieving the effective and target specific delivery of pharmaceutical payloads. Adoption of nanotechnological approaches in delivering therapeutic molecules to target cells ensures not only reduced immune response and toxicity, but increases the stability of therapeutic entities in the systemic circulation that averts their degradation and as such increased extravasations and accumulation via enhanced permeation and the retention (EPR) effect in target tissues. Additionally, nanoparticle (NP)-induced ER stress, which enhances apoptosis and autophagy, has been utilized as a combative strategy in the treatment of cancerous cells. As nanoparticles-based avenues have been capitalized to achieve better efficacy of the new genera of therapeutics with enhanced specificity and safety, the present study is aimed at providing the fundamentals of BC, nanotechnological modules (organic, inorganic, and hybrid) employed in delivering different therapeutic molecules, and mechanistic insights of nano-ER stress induced apoptosis and autophagy with a perspective of exploring this avenue for use in the nano-toxicological studies. Furthermore, the current scenario of USA FDA approved nano-formulations and the future perspective of nanotechnological based interventions to overcome the existing challenges are also discussed.
Collapse
Affiliation(s)
- Safikur Rahman
- Department of Botany, Munshi Singh College, BR Ambedkar Bihar University, Muzaffarpur 845401, India;
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Korea;
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea;
| | - Tasduq S. Abdullah
- Council of Scientific and Industrial Research–Indian Institute of Integrative Medicine (CSIR–IIIM), Jammu 180001, India;
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), P.O. Box 80141, Jeddah 21589, Saudi Arabia
- Correspondence: (I.A.R.); (A.T.J.)
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India
- Correspondence: (I.A.R.); (A.T.J.)
| |
Collapse
|
20
|
Hesari M, Mohammadi P, Khademi F, Shackebaei D, Momtaz S, Moasefi N, Farzaei MH, Abdollahi M. Current Advances in the Use of Nanophytomedicine Therapies for Human Cardiovascular Diseases. Int J Nanomedicine 2021; 16:3293-3315. [PMID: 34007178 PMCID: PMC8123960 DOI: 10.2147/ijn.s295508] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Considering the high prevalence of cardiovascular diseases (CVDs), the primary cause of death during the last several decades, it is necessary to develop proper strategies for the prevention and treatment of CVDs. Given the excessive side effects of current therapies, alternative therapeutic approaches like medicinal plants and natural products are preferred. Lower toxicity, chemical diversity, cost-effectiveness, and proven therapeutic potentials make natural products superior compared to other products. Nanoformulation methods improve the solubility, bioavailability, circulation time, surface area-to-volume ratio, systemic adverse side effects, and drug delivery efficiency of these medications. This study intended to review the functionality of the most recent nanoformulated medicinal plants and/or natural products against various cardiovascular conditions such as hypertension, atherosclerosis, thrombosis, and myocardial infarction. Literature review revealed that curcumin, quercetin, and resveratrol were the most applied natural products, respectively. Combination therapy, conjugation, or fabrication of nanoparticles and nanocarriers improved the applications and therapeutic efficacy of herbal- or natural-based nanoformulations. In the context of CVDs prevention and/or treatment, available data suggest that natural-based nanoformulations are considerably efficient, alone or in blend with other herbal/synthetic medicines. However, clinical trials are mandatory to elucidate the safety, cardioprotective effect, and mechanism of actions of nanophytomedicines.
Collapse
Affiliation(s)
- Mahvash Hesari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Khademi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Dareuosh Shackebaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Gastrointestinal Pharmacology Interest Group, Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Narges Moasefi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Zhang T, Fu C, Alradwan I, Yen T, Lip H, Cai P, Rauth AM, Zhang L, Wu XY. Targeting Signaling Pathways of Hyaluronic Acid and Integrin Receptors by Synergistic Combination Nanocomposites Inhibits Systemic Metastases and Primary Triple Negative Breast Cancer. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Tian Zhang
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy University of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Chaoping Fu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy University of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Ibrahim Alradwan
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy University of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - TinYo Yen
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy University of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - HoYin Lip
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy University of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Ping Cai
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy University of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Andrew M. Rauth
- Departments of Medical Biophysics and Radiation Oncology University of Toronto 610 University Ave Toronto Ontario M5G 2M9 Canada
| | - Liming Zhang
- DSAPM Lab and PCFM Lab, School of Materials Science and Engineering Sun Yat‐sen University Guangzhou 510275 P. R. China
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy University of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| |
Collapse
|
22
|
Combined inhibition of CD73 and ZEB1 by Arg-Gly-Asp (RGD)-targeted nanoparticles inhibits tumor growth. Colloids Surf B Biointerfaces 2021; 197:111421. [DOI: 10.1016/j.colsurfb.2020.111421] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 12/15/2022]
|
23
|
Kang C, Han P, Lee JS, Lee D, Kim D. Anchor, Spacer, and Ligand-Modified Engineered Exosomes for Trackable Targeted Therapy. Bioconjug Chem 2020; 31:2541-2552. [PMID: 33115231 DOI: 10.1021/acs.bioconjchem.0c00483] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Exosomes have been widely demonstrated as an effective anticancer therapeutic moiety. However, their clinical translation has been limited by the requirement of prohibitively high therapeutic doses due to their lack of specificity in delivery and, consequently, short systemic half-life. To overcome these challenges, we engineered a platform for modifying exosomes with an active targeting modality composed of membrane Anchor (BODIPY)-Spacer (PEG)-targeting Ligands (cyclic RGD peptide) (ASL). Herein, we show that the intramembrane incorporation of a trackable, targeting system renders ASL exosomes (AExs) a modular platform. AExs significantly overcome challenges associated with exosome modification, including potential damage for functionalization, or destabilizing interactions between dyes and drugs. ASL-modification not only enhanced stability in imparting active targeting but also introduced a built-in bioimaging modality. Our studies show that AExs target B16F10 melanoma tumor sites by the specific interaction of cyclic RGD and integrin. Doxorubicin encapsulated AExs (dAExs) significantly inhibited the growth of melanoma in vitro and in vivo. Thus, we conclude that ASL-modification allows exosomes to be transformed into a novel therapeutic vehicle uniquely integrating in vivo tracking and robust targeting with drug delivery. We anticipate that the therapeutic, targeting, and diagnostic modularity provided by ASL will potentiate translational applications of exosome-based vehicles beyond anticancer therapy.
Collapse
Affiliation(s)
- Changsun Kang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Patrick Han
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, United States
| | - Jung S Lee
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, United States
| | - Dongwon Lee
- Department of Polymer·Nano Science and Technology, Chonbuk National University, Jeonju, South Korea, 54896
| | - Dongin Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| |
Collapse
|
24
|
Alshehri S, Fan W, Zhang W, Garrison JC. In Vitro Evaluation and Biodistribution Studies of HPMA Copolymers Targeting the Gastrin Releasing Peptide Receptor in Prostate Cancer. Pharm Res 2020; 37:229. [PMID: 33098043 DOI: 10.1007/s11095-020-02952-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE The development of diagnostic and therapeutic agents utilizing small peptides (e.g., bombesin (BBN)) to target the overexpression of the gastrin-releasing peptide receptor (GRPR) in cancers has been widely investigated. Herein, we examine the capabilities of BBN-modified HPMA copolymers to target the GRPR. METHODS Four positive, four negative, and two zwitterionic BBN HPMA copolymer conjugates of varying peptide content and charge were synthesized. In vitro and in vivo studies were conducted in a GRPR-overexpressing prostate cancer cell line (PC-3) and a normal CF-1 mouse model, respectively. RESULTS Cellular uptake of the conjugates were found to be charge and BBN density dependent. The positively-charged conjugates illustrated a direct relationship between the extent of cellular internalization, ranging from 0.7 to 20%, and BBN-incorporation density. The negative and zwitterionic conjugates showed low PC-3 uptake values. Blocking studies confirmed the GRPR-targeting effect of the positively-charged constructs. In vivo studies of the positively-charged copolymers resulted in rapid blood clearance by the mononuclear phagocyte system (MPS)-associated tissues (e.g., liver and spleen). CONCLUSION Positively-charged BBN-HPMA copolymer conjugates demonstrated good GRPR-targeting and internalization in vitro. However, the impact of peptide density and charge on in vivo MPS recognition are parameters that must be optimized in future agent development.
Collapse
Affiliation(s)
- Sameer Alshehri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska, 68198, USA
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska, 68198, USA
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Wei Fan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska, 68198, USA
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Wenting Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska, 68198, USA
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Jered C Garrison
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska, 68198, USA.
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska, 68198, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, Nebraska, 68198, USA.
- Eppley Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska, 68198, USA.
| |
Collapse
|
25
|
Ji X, Han T, Kang N, Huang S, Liu Y. Preparation of RGD4C fused anti-TNFα nanobody and inhibitory activity on triple-negative breast cancer in vivo. Life Sci 2020; 260:118274. [PMID: 32827545 DOI: 10.1016/j.lfs.2020.118274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 01/13/2023]
Abstract
AIMS Triple-negative breast cancer (TNBC) is not sensitive to current endocrine treatments, so new treatment strategies need to be explored. Based on previous antitumour studies on anti-TNFα nanobody, we designed a novel fusion nanobody to enhance antitumour activity of the anti-TNFα nanobody in TNBC. MAIN METHODS The RGD4C contains RGD sequence, which is the smallest recognition unit binding to the αvβ3 receptor on tumour cell membranes and involved in tumour cell adhesion, proliferation, and metastasis. RGD4C was fused to anti-TNFα nanobody to investigate the antitumour activity in vitro and in vivo. KEY FINDINGS The antitumour effects of fusion nanobody V-L-R-H could effectively bind to αvβ3 and inhibit cell migration and proliferation of MDA-MB-231, which had satisfying purification efficiency and approving antigen or receptor binding activity. V-L-R-H could inhibit the TNFα-mediated PI3K/AKT/NF-κB signal pathway and integrin αvβ3 correlative FAK focal adhesion signal pathway. Mouse xenograft tumour experiments showed that the V-L-R-H could inhibit tumour proliferation and metastasis; reduce the TNFα, HIFα, Ki67, and CD31 concentrations in tumour; and inhibit the process of epithelial-mesenchymal transition. SIGNIFICANCE The fusion nanobody enhanced antitumour activity of the anti-TNFα nanobody on TNBC. It provided a reference for the design of dual functional fusion proteins and development of tumour treatment strategies of antagonistic TNFα and αvβ3, and a new therapeutic strategy and research direction for the treatment of TNBC.
Collapse
Affiliation(s)
- Xuemei Ji
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Tianzhen Han
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Nannan Kang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Song Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yu Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
26
|
Mahmoudi R, Ashraf Mirahmadi-Babaheidri S, Delaviz H, Fouani MH, Alipour M, Jafari Barmak M, Christiansen G, Bardania H. RGD peptide-mediated liposomal curcumin targeted delivery to breast cancer cells. J Biomater Appl 2020; 35:743-753. [PMID: 32807016 DOI: 10.1177/0885328220949367] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this study, turmeric's active ingredient (Curcumin) was encapsulated into RGD modified Liposomes (RGD-Lip-Cur) its cytotoxic effect on the breast cancer cell line (MCF-7) was evaluated by MTT, flow cytometry and Caspase assay. Liposomes were characterized using transmission electron microscopy (TEM). Results demonstrated that the liposomes were spherical in shape, ranging from 70 to 100 nm. MTT assay revealed that RGD-Lip-Cur had a significant cytotoxic effect on MCF-7 cells at concentrations of 32, 16 and 4 μg/ml compared to Lip-Cur (P < 0.05) and curcumin (P < 0.01). The apoptosis assay demonstrated that RGD-Lip-Cur induces the apoptosis in MCF-7 cells (39.6% vs 40.2% for initial and secondary apoptosis) significantly more than Lip-Cur (67.7% vs 9.16% for initial and secondary apoptosis) and free curcumin (7.84% vs 38.8% for initial and secondary apoptosis). Moreover, caspase assay showed that RGD-Lip-Cur activates caspase 3/7 compared to Lip-Cur (P < 0.05) and free curcumin (P < 0.01). The RGD-Lip-Cur was similar to the control group and had no significant cytotoxicity effect. It is concluded that RGD-Lip-Cur as a novel carrier have high cytotoxicity effect on breast cancer cell line (MCF-7).
Collapse
Affiliation(s)
- Reza Mahmoudi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Hamdollah Delaviz
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohamad Hassan Fouani
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohsen Alipour
- Department of Advanced Medical Sciences & Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mehrzad Jafari Barmak
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Medicinal Plant Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Clinical Research Development Unit, Imamsajad Hospital, Yasuj University of Medical Sciences, Yasuj, Iran
| |
Collapse
|
27
|
Bao H, Zheng N, Li Z, Zhi Y. Synergistic Effect of Tangeretin and Atorvastatin for Colon Cancer Combination Therapy: Targeted Delivery of These Dual Drugs Using RGD Peptide Decorated Nanocarriers. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3057-3068. [PMID: 32801644 PMCID: PMC7397562 DOI: 10.2147/dddt.s256636] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022]
Abstract
Purpose Colorectal cancer (CRC) is the third most frequently diagnosed cancer and the fourth leading cause of cancer death over the world. Nano-sized drug delivery systems are used for the treatment of cancers. The aim of this study was to develop a tangeretin (TAGE) and atorvastatin (ATST) combined nano-system decorated with RGD (RGD-ATST/TAGE CNPs) for colon cancer combination therapy. Materials and Methods In this study, cyclized arginine-glycine-aspartic acid sequences (RGD) contained ligand was synthesized by conjugating cyclo (Arg-Gly-Asp-d-Phe-Lys) (cRGDfK) with D-α-tocopheryl succinate dichloromethane (TOSD) using polyethylene glycol (PEG) as a linker to obtain cRGDfK-PEG-TOSD. ATST and TAGE combined nano-systems: RGD-ATST/TAGE CNPs were prepared. The combination effects as well as antitumor effects of these two agents were evaluated on colon cancer cells and mice bearing cancer models. Results Drug entrapment efficiencies of nano-systems were high (around 90%), suggesting the good loading capacity. The release profiles of ATST or TAGE from RGD-ATST/TAGE CNPs followed Higuchi model. The RGD-decorated nano-system showed more obvious cytotoxicity on HT-29 cells than the undecorated nano-system, but no obvious difference was found on normal CCD-18 cells. The strongest synergism was observed when the weight ratio of ATST to TAGE was 1:1. In vivo biodistribution of RGD-ATST/TAGE CNPs in the tumor site is high and prominently inhibited the in vivo tumor growth. Conclusion The results demonstrated that RGD-ATST/TAGE CNPs showed the most significant synergistic therapeutic efficacy, exhibited no significant toxicity to major organs and tissues, and body weight of the treated mice was stable. Therefore, the combination nano-system is a promising platform for colon cancer therapy.
Collapse
Affiliation(s)
- He Bao
- Department of Pharmacy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| | - Nanbo Zheng
- Department of Pharmacy, Xi'an Central Hospital, Xi'an 710003, People's Republic of China
| | - Zhuanting Li
- Department of Pharmacy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| | - Yuan Zhi
- Department of Pharmacy, Xi'an Hospital of Traditional Chinese Medicine, Xi'an 710021, Shaanxi, People's Republic of China
| |
Collapse
|
28
|
Leach A, Smyth P, Ferguson L, Steven J, Greene MK, Branco CM, McCann AP, Porter A, Barelle CJ, Scott CJ. Anti-DLL4 VNAR targeted nanoparticles for targeting of both tumour and tumour associated vasculature. NANOSCALE 2020; 12:14751-14763. [PMID: 32626858 DOI: 10.1039/d0nr02962a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Whilst there is an extensive body of preclinical nanomedicine research, translation to clinical settings has been slow. Here we present a novel approach to the targeted nanoparticle (NP) concept: utilizing both a novel targeting ligand, VNAR (Variable New Antigen Receptor), a shark-derived single chain binding domain, and an under-investigated target in delta-like ligand 4 (DLL4). We describe the development of an anti-DLL4 VNAR and the site-specific conjugation of this to poly(lactic-co-glycolic) acid PEGylated NPs using surface maleimide functional groups. These nanoconjugates were shown to specifically bind DLL4 with high affinity and were preferentially internalized by DLL4-expressing pancreatic cancer cell lines and endothelial cells. Furthermore, a distinct anti-angiogenic effect endowed by the anti-DLL4 VNAR was evident in in vitro tubulogenic assays. Taken together these findings highlight the potential of anti-DLL4 targeted polymeric NPs as a novel therapeutic approach in pancreatic cancer.
Collapse
Affiliation(s)
- Adam Leach
- The Patrick G. Johnston Centre for Cancer Research, School of Medicine, Queen's University Belfast, Belfast, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Endothelial Cell Targeting by cRGD-Functionalized Polymeric Nanoparticles under Static and Flow Conditions. NANOMATERIALS 2020; 10:nano10071353. [PMID: 32664364 PMCID: PMC7407316 DOI: 10.3390/nano10071353] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Since αvβ3 integrin is a key component of angiogenesis in health and disease, Arg-Gly-Asp (RGD) peptide-functionalized nanocarriers have been investigated as vehicles for targeted delivery of drugs to the αvβ3 integrin-overexpressing neovasculature of tumors. In this work, PEGylated nanoparticles (NPs) based on poly(lactic-co-glycolic acid) (PLGA) functionalized with cyclic-RGD (cRGD), were evaluated as nanocarriers for the targeting of angiogenic endothelium. For this purpose, NPs (~300 nm) functionalized with cRGD with different surface densities were prepared by maleimide-thiol chemistry and their interactions with human umbilical vein endothelial cells (HUVECs) were evaluated under different conditions using flow cytometry and microscopy. The cell association of cRGD-NPs under static conditions was time-, concentration- and cRGD density-dependent. The interactions between HUVECs and cRGD-NPs dispersed in cell culture medium under flow conditions were also time- and cRGD density-dependent. When washed red blood cells (RBCs) were added to the medium, a 3 to 8-fold increase in NPs association to HUVECs was observed. Moreover, experiments conducted under flow in the presence of RBC at physiologic hematocrit and shear rate, are a step forward in the prediction of in vivo cell–particle association. This approach has the potential to assist development and high-throughput screening of new endothelium-targeted nanocarriers.
Collapse
|
30
|
Garanti T, Alhnan MA, Wan KW. RGD-decorated solid lipid nanoparticles enhance tumor targeting, penetration and anticancer effect of asiatic acid. Nanomedicine (Lond) 2020; 15:1567-1583. [DOI: 10.2217/nnm-2020-0035] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: Asiatic acid (AA) is a promising anticancer agent, however, its delivery to glioblastoma is a major challenge. This work investigates the beneficial therapeutic efficacy of RGD-conjugated solid lipid nanoparticles (RGD-SLNs) for the selective targeting of AA to gliblastoma. Materials & methods: AA-containing RGD-SLNs were prepared using two different PEG-linker size. Targetability and efficacy were tested using monolayer cells and spheroid tumor models. Results: RGD-SLNs significantly improved cytotoxicity of AA against U87-MG monolayer cells and enhanced cellular uptake compared with non-RGD-containing SLNs. In spheroid models, AA-containing RGD-SLNs showed superior control in tumor growth, improved cytotoxicity and enhanced spheroid penetration when compared with AA alone or non-RGD-containing SLNs. Conclusion: This study illustrates the potential of AA-loaded RGD-SLNs as efficacious target-specific treatment for glioblastoma.
Collapse
Affiliation(s)
- Tanem Garanti
- Faculty of Pharmacy, Cyprus International University, Haspolat, Nicosia, 99258, Cyprus via Mersin 10, Turkey
| | - Mohamed A Alhnan
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, London, UK
| | - Ka-Wai Wan
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston, Lancashire, PR1 2HE, UK
| |
Collapse
|
31
|
Ding F, Fu J, Tao C, Yu Y, He X, Gao Y, Zhang Y. Recent Advances of Chitosan and its Derivatives in Biomedical Applications. Curr Med Chem 2020; 27:3023-3045. [DOI: 10.2174/0929867326666190405151538] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 12/29/2022]
Abstract
Chitosan is the second-most abundant natural polysaccharide. It has unique characteristics,
such as biodegradability, biocompatibility, and non-toxicity. Due to the existence of its free amine
group and hydroxyl groups on its backbone chain, chitosan can undergo further chemical modifications
to generate Chitosan Derivatives (CDs) that permit additional biomedical functionality. Chitosan
and CDs can be fabricated into various forms, including Nanoparticles (NPs), micelles, hydrogels,
nanocomposites and nano-chelates. For these reasons, chitosan and CDs have found a tremendous
variety of biomedical applications in recent years. This paper mainly presents the prominent
applications of chitosan and CDs for cancer therapy/diagnosis, molecule biosensing, viral infection,
and tissue engineering over the past five years. Moreover, future research directions on chitosan are
also considered.
Collapse
Affiliation(s)
- Fei Ding
- Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, 430056, China
| | - Jiawei Fu
- Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, 430056, China
| | - Chuang Tao
- Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, 430056, China
| | - Yanhua Yu
- Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, 430056, China
| | - Xianran He
- Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, 430056, China
| | - Yangguang Gao
- Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, 430056, China
| | - Yongmin Zhang
- Institut Parisien de Chimie Moléculaire, CNRS UMR 8232, Sorbonne Université, 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
32
|
Bidram E, Esmaeili Y, Ranji-Burachaloo H, Al-Zaubai N, Zarrabi A, Stewart A, Dunstan DE. A concise review on cancer treatment methods and delivery systems. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101350] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
33
|
Sieber S, Grossen P, Bussmann J, Campbell F, Kros A, Witzigmann D, Huwyler J. Zebrafish as a preclinical in vivo screening model for nanomedicines. Adv Drug Deliv Rev 2019; 151-152:152-168. [PMID: 30615917 DOI: 10.1016/j.addr.2019.01.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/23/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022]
Abstract
The interactions of nanomedicines with biological environments is heavily influenced by their physicochemical properties. Formulation design and optimization are therefore key steps towards successful nanomedicine development. Unfortunately, detailed assessment of nanomedicine formulations, at a macromolecular level, in rodents is severely limited by the restricted imaging possibilities within these animals. Moreover, rodent in vivo studies are time consuming and expensive, limiting the number of formulations that can be practically assessed in any one study. Consequently, screening and optimisation of nanomedicine formulations is most commonly performed in surrogate biological model systems, such as human-derived cell cultures. However, despite the time and cost advantages of classical in vitro models, these artificial systems fail to reflect and mimic the complex biological situation a nanomedicine will encounter in vivo. This has acutely hampered the selection of potentially successful nanomedicines for subsequent rodent in vivo studies. Recently, zebrafish have emerged as a promising in vivo model, within nanomedicine development pipelines, by offering opportunities to quickly screen nanomedicines under in vivo conditions and in a cost-effective manner so as to bridge the current gap between in vitro and rodent studies. In this review, we outline several advantageous features of the zebrafish model, such as biological conservation, imaging modalities, availability of genetic tools and disease models, as well as their various applications in nanomedicine development. Critical experimental parameters are discussed and the most beneficial applications of the zebrafish model, in the context of nanomedicine development, are highlighted.
Collapse
Affiliation(s)
- Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jeroen Bussmann
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Alexander Kros
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Department of Biochemistry and Molecular Biology, University of British Columbia, Health Sciences Mall, Vancouver, British Columbia, Canada..
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
34
|
Thakur V, Kutty RV. Recent advances in nanotheranostics for triple negative breast cancer treatment. J Exp Clin Cancer Res 2019; 38:430. [PMID: 31661003 PMCID: PMC6819447 DOI: 10.1186/s13046-019-1443-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most complex and aggressive type of breast cancer encountered world widely in women. Absence of hormonal receptors on breast cancer cells necessitates the chemotherapy as the only treatment regime. High propensity to metastasize and relapse in addition to poor prognosis and survival motivated the oncologist, nano-medical scientist to develop novel and efficient nanotherapies to solve such a big TNBC challenge. Recently, the focus for enhanced availability, targeted cellular uptake with minimal toxicity is achieved by nano-carriers. These smart nano-carriers carrying all the necessary arsenals (drugs, tracking probe, and ligand) designed in such a way that specifically targets the TNBC cells at site. Articulating the targeted delivery system with multifunctional molecules for high specificity, tracking, diagnosis, and treatment emerged as theranostic approach. In this review, in addition to classical treatment modalities, recent advances in nanotheranostics for early and effective diagnostic and treatment is discussed. This review highlighted the recently FDA approved immunotherapy and all the ongoing clinical trials for TNBC, in addition to nanoparticle assisted immunotherapy. Futuristic but realistic advancements in artificial intelligence (AI) and machine learning not only improve early diagnosis but also assist clinicians for their workup in TNBC. The novel concept of Nanoparticles induced endothelial leakiness (NanoEL) as a way of tumor invasion is also discussed in addition to classical EPR effect. This review intends to provide basic insight and understanding of the novel nano-therapeutic modalities in TNBC diagnosis and treatment and to sensitize the readers for continue designing the novel nanomedicine. This is the first time that designing nanoparticles with stoichiometric definable number of antibodies per nanoparticle now represents the next level of precision by design in nanomedicine.
Collapse
Affiliation(s)
- Vikram Thakur
- Department of Virology, Postgraduate Institute of Medical Education and Research, PGIMER, Chandigarh, 160012 India
| | - Rajaletchumy Veloo Kutty
- Faculty of Chemical and Process Engineering Technology, College of Engineering Technology,University Malaysia Pahang, Tun Razak Highway, 26300 Kuantan, Pahang Malaysia
- Center of Excellence for Advanced Research in Fluid Flow, University Malaysia Pahang, 26300, Kuantan, Pahang Malaysia
| |
Collapse
|
35
|
Rajabi M, Adeyeye M, Mousa SA. Peptide-Conjugated Nanoparticles as Targeted Anti-angiogenesis Therapeutic and Diagnostic in Cancer. Curr Med Chem 2019; 26:5664-5683. [DOI: 10.2174/0929867326666190620100800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/11/2019] [Accepted: 03/21/2019] [Indexed: 12/25/2022]
Abstract
:Targeting angiogenesis in the microenvironment of a tumor can enable suppression of tumor angiogenesis and delivery of anticancer drugs into the tumor. Anti-angiogenesis targeted delivery systems utilizing passive targeting such as Enhanced Permeability and Retention (EPR) and specific receptor-mediated targeting (active targeting) should result in tumor-specific targeting. One targeted anti-angiogenesis approach uses peptides conjugated to nanoparticles, which can be loaded with anticancer agents. Anti-angiogenesis agents can suppress tumor angiogenesis and thereby affect tumor growth progression (tumor growth arrest), which may be further reduced with the targetdelivered anticancer agent. This review provides an update of tumor vascular targeting for therapeutic and diagnostic applications, with conventional or long-circulating nanoparticles decorated with peptides that target neovascularization (anti-angiogenesis) in the tumor microenvironment.
Collapse
Affiliation(s)
- Mehdi Rajabi
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Mary Adeyeye
- Department of Chemistry, University of Albany, State University of New York, Albany, NY 12222, United States
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| |
Collapse
|
36
|
Zhang T, Lip H, He C, Cai P, Wang Z, Henderson JT, Rauth AM, Wu XY. Multitargeted Nanoparticles Deliver Synergistic Drugs across the Blood-Brain Barrier to Brain Metastases of Triple Negative Breast Cancer Cells and Tumor-Associated Macrophages. Adv Healthc Mater 2019; 8:e1900543. [PMID: 31348614 DOI: 10.1002/adhm.201900543] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/12/2019] [Indexed: 12/14/2022]
Abstract
Patients with brain metastases of triple negative breast cancer (TNBC) have a poor prognosis owing to the lack of targeted therapies, the aggressive nature of TNBC, and the presence of the blood-brain barrier (BBB) that blocks penetration of most drugs. Additionally, infiltration of tumor-associated macrophages (TAMs) promotes tumor progression. Here, a terpolymer-lipid hybrid nanoparticle (TPLN) system is designed with multiple targeting moieties to first undergo synchronized BBB crossing and then actively target TNBC cells and TAMs in microlesions of brain metastases. In vitro and in vivo studies demonstrate that covalently bound polysorbate 80 in the terpolymer enables the low-density lipoprotein receptor-mediated BBB crossing and TAM-targetability of the TPLN. Conjugation of cyclic internalizing peptide (iRGD) enhances cellular uptake, cytotoxicity, and drug delivery to brain metastases of integrin-overexpressing TNBC cells. iRGD-TPLN with coloaded doxorubicin (DOX) and mitomycin C (MMC) (iRGD-DMTPLN) exhibits higher efficacy in reducing metastatic burden and TAMs than nontargeted DMTPLN or a free DOX/MMC combination. iRGD-DMTPLN treatment reduces metastatic burden by 6-fold and 19-fold and increases host median survival by 1.3-fold and 1.6-fold compared to DMTPLN or free DOX/MMC treatments, respectively. These findings suggest that iRGD-DMTPLN is a promising multitargeted drug delivery system for the treatment of integrin-overexpressing brain metastases of TNBC.
Collapse
Affiliation(s)
- Tian Zhang
- Advanced Pharmaceutics and Drug Delivery LaboratoryLeslie Dan Faculty of PharmacyUniversity of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Hoyin Lip
- Advanced Pharmaceutics and Drug Delivery LaboratoryLeslie Dan Faculty of PharmacyUniversity of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Chunsheng He
- Advanced Pharmaceutics and Drug Delivery LaboratoryLeslie Dan Faculty of PharmacyUniversity of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Ping Cai
- Advanced Pharmaceutics and Drug Delivery LaboratoryLeslie Dan Faculty of PharmacyUniversity of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Zhigao Wang
- Advanced Pharmaceutics and Drug Delivery LaboratoryLeslie Dan Faculty of PharmacyUniversity of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Jeffrey T. Henderson
- Advanced Pharmaceutics and Drug Delivery LaboratoryLeslie Dan Faculty of PharmacyUniversity of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Andrew M. Rauth
- Departments of Medical Biophysics and Radiation OncologyUniversity of Toronto 610 University Ave Toronto Ontario M5G 2M9 Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery LaboratoryLeslie Dan Faculty of PharmacyUniversity of Toronto 144 College Street Toronto Ontario M5S 3M2 Canada
| |
Collapse
|
37
|
Ahmad A, Khan F, Mishra RK, Khan R. Precision Cancer Nanotherapy: Evolving Role of Multifunctional Nanoparticles for Cancer Active Targeting. J Med Chem 2019; 62:10475-10496. [DOI: 10.1021/acs.jmedchem.9b00511] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Anas Ahmad
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali 160062, Punjab, India
| | - Farheen Khan
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali 160062, Punjab, India
| | - Rakesh Kumar Mishra
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali 160062, Punjab, India
| | - Rehan Khan
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali 160062, Punjab, India
| |
Collapse
|
38
|
Witzigmann D, Uhl P, Sieber S, Kaufman C, Einfalt T, Schöneweis K, Grossen P, Buck J, Ni Y, Schenk SH, Hussner J, Meyer Zu Schwabedissen HE, Québatte G, Mier W, Urban S, Huwyler J. Optimization-by-design of hepatotropic lipid nanoparticles targeting the sodium-taurocholate cotransporting polypeptide. eLife 2019; 8:42276. [PMID: 31333191 PMCID: PMC6682401 DOI: 10.7554/elife.42276] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Active targeting and specific drug delivery to parenchymal liver cells is a promising strategy to treat various liver disorders. Here, we modified synthetic lipid-based nanoparticles with targeting peptides derived from the hepatitis B virus large envelope protein (HBVpreS) to specifically target the sodium-taurocholate cotransporting polypeptide (NTCP; SLC10A1) on the sinusoidal membrane of hepatocytes. Physicochemical properties of targeted nanoparticles were optimized and NTCP-specific, ligand-dependent binding and internalization was confirmed in vitro. The pharmacokinetics and targeting capacity of selected lead formulations was investigated in vivo using the emerging zebrafish screening model. Liposomal nanoparticles modified with 0.25 mol% of a short myristoylated HBV derived peptide, that is Myr-HBVpreS2-31, showed an optimal balance between systemic circulation, avoidance of blood clearance, and targeting capacity. Pronounced liver enrichment, active NTCP-mediated targeting of hepatocytes and efficient cellular internalization were confirmed in mice by 111In gamma scintigraphy and fluorescence microscopy demonstrating the potential use of our hepatotropic, ligand-modified nanoparticles.
Collapse
Affiliation(s)
- Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Philipp Uhl
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Christina Kaufman
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, INF, Heidelberg, Germany
| | - Tomaz Einfalt
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Katrin Schöneweis
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, INF, Heidelberg, Germany
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jonas Buck
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Yi Ni
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, INF, Heidelberg, Germany
| | - Susanne H Schenk
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Janine Hussner
- Division of Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | | | - Gabriela Québatte
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, INF, Heidelberg, Germany
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
39
|
Zheng G, Zheng M, Yang B, Fu H, Li Y. Improving breast cancer therapy using doxorubicin loaded solid lipid nanoparticles: Synthesis of a novel arginine-glycine-aspartic tripeptide conjugated, pH sensitive lipid and evaluation of the nanomedicine in vitro and in vivo. Biomed Pharmacother 2019; 116:109006. [PMID: 31152925 DOI: 10.1016/j.biopha.2019.109006] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/03/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the leading cause of cancer mortality in women worldwide. To overcome the toxic side effects and multidrug resistance (MDR) during doxorubicin (DOX) chemotherapy, an arginine-glycine-aspartic (RGD) tripeptide modified, pH-sensitive solid lipid nanoparticles (SLNs) is employed in this study. In this study, a RGD conjugated, pH sensitive lipid was synthesized using glycerin monostearate (GMS) and adipic acid dihydrazide (HZ) as lipid materials and named RGD-HZ-GMS. RGD-HZ-GMS was applied to encapsulate DOX to construct a RGD modified, DOX loaded SLNs (RGD-DOX-SLNs). To evaluate the anticancer effect of RGD-DOX-SLNs, breast cancer cell line (MCF-7 cells) and DOX resistant cell line (MCF-7/ADR cells) were used. in vivo tumor suspension and toxicity effects were evaluated on mice bearing MCF-7/ADR cells breast cancer model. RGD-DOX-SLNs had a uniformly spherical shape. The mean particle size and zeta potential of the RGD-DOX-SLNs was 96.3 nm and 35.6 mV, respectively. RGD-DOX-SLNs showed 5.58 fold higher area under the plasma concentration - time curve (AUC) compared with DOX solution. Terminal half life (T1/2) and peak concentration (Cmax) of RGD-DOX-SLNs was 10.85 h and 39.12 ± 2.71 L/kg/h. in vitro and in vivo antitumor results indicate that RGD-DOX-SLNs might be a promising novel lipid carrier which could improve breast cancer therapy.
Collapse
Affiliation(s)
- Gang Zheng
- Department of Surgical Ward 2, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, PR China
| | - Meizhu Zheng
- Department of Surgical Ward 2, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, PR China
| | - Ben Yang
- Department of Surgical Ward 2, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, PR China
| | - Hui Fu
- Department of Surgical Ward 2, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, PR China
| | - Yongqing Li
- Department of Surgical Ward 1, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, PR China.
| |
Collapse
|
40
|
Gajbhiye KR, Gajbhiye V, Siddiqui IA, Gajbhiye JM. cRGD functionalised nanocarriers for targeted delivery of bioactives. J Drug Target 2018; 27:111-124. [PMID: 29737883 DOI: 10.1080/1061186x.2018.1473409] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The integrins αvβ3 play a very imperative role in angiogenesis and are overexpressed in endothelial cells of the tumour. Recent years have witnessed huge exploration in the field of αvβ3 integrin-mediated bioactive targeting for treatment of cancer. In these studies, the cRGD peptide has been employed extensively owing to their binding capacity to the αvβ3 integrin. Principally, RGD-based approaches comprise of antagonist molecules of the RGD sequence, drug-RGD conjugates, and most importantly tethering of the nanocarrier surface with the RGD peptide as targeting ligand. Targeting tumour vasculature or cells via cRGD conjugated nanocarriers have emerged as a promising technique for delivering chemotherapeutic drugs and imaging agents for cancer theranostics. In this review, primary emphasis has been given on the application of cRGD-anchored nanocarriers for targeted delivery of drugs, imaging agents, etc. for tumour therapy.
Collapse
Affiliation(s)
- K R Gajbhiye
- a Division of Organic Chemistry , CSIR-National Chemical Laboratory , Pune , India
| | - V Gajbhiye
- b Nanobioscience , Agharkar Research Institute , Pune , India
| | - Imtiaz A Siddiqui
- c Department of Dermatology , University of Wisconsin , Madison , WI , USA
| | - J M Gajbhiye
- a Division of Organic Chemistry , CSIR-National Chemical Laboratory , Pune , India
| |
Collapse
|
41
|
Zhang RX, Li J, Zhang T, Amini MA, He C, Lu B, Ahmed T, Lip H, Rauth AM, Wu XY. Importance of integrating nanotechnology with pharmacology and physiology for innovative drug delivery and therapy - an illustration with firsthand examples. Acta Pharmacol Sin 2018; 39:825-844. [PMID: 29698389 DOI: 10.1038/aps.2018.33] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/19/2018] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology has been applied extensively in drug delivery to improve the therapeutic outcomes of various diseases. Tremendous efforts have been focused on the development of novel nanoparticles and delineation of the physicochemical properties of nanoparticles in relation to their biological fate and functions. However, in the design and evaluation of these nanotechnology-based drug delivery systems, the pharmacology of delivered drugs and the (patho-)physiology of the host have received less attention. In this review, we discuss important pharmacological mechanisms, physiological characteristics, and pathological factors that have been integrated into the design of nanotechnology-enabled drug delivery systems and therapies. Firsthand examples are presented to illustrate the principles and advantages of such integrative design strategies for cancer treatment by exploiting 1) intracellular synergistic interactions of drug-drug and drug-nanomaterial combinations to overcome multidrug-resistant cancer, 2) the blood flow direction of the circulatory system to maximize drug delivery to the tumor neovasculature and cells overexpressing integrin receptors for lung metastases, 3) endogenous lipoproteins to decorate nanocarriers and transport them across the blood-brain barrier for brain metastases, and 4) distinct pathological factors in the tumor microenvironment to develop pH- and oxidative stress-responsive hybrid manganese dioxide nanoparticles for enhanced radiotherapy. Regarding the application in diabetes management, a nanotechnology-enabled closed-loop insulin delivery system was devised to provide dynamic insulin release at a physiologically relevant time scale and glucose levels. These examples, together with other research results, suggest that utilization of the interplay of pharmacology, (patho-)physiology and nanotechnology is a facile approach to develop innovative drug delivery systems and therapies with high efficiency and translational potential.
Collapse
|
42
|
Fu S, Xu X, Ma Y, Zhang S, Zhang S. RGD peptide-based non-viral gene delivery vectors targeting integrin α vβ 3 for cancer therapy. J Drug Target 2018; 27:1-11. [PMID: 29564914 DOI: 10.1080/1061186x.2018.1455841] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Integrin αvβ3 is restrictedly expressed on angiogenic blood vessels and tumour cells. It plays a key role in angiogenesis for tumour growth and metastasis. RGD peptide can specifically recognise the integrin αvβ3, which serves as targeted molecular for anti-angiogenesis strategies. Therefore, the targeted delivery of therapeutics by RGD peptide-based non-viral vectors to tumour vasculature and tumour cells is recognised as a promising approach for treating cancer. In this review, we illustrate the interaction between RGD peptide and integrin αvβ3 from different perspectives. Meanwhile, four types of RGD peptide-based non-viral gene delivery vectors for cancer therapy, including RGD-based cationic polymers, lipids, peptides and hybrid systems, are summarised. The aim is to particularly highlight the enhanced therapeutic effects and specific targeting ability exhibited by these vectors for cancer gene therapy both in vitro and in vivo.
Collapse
Affiliation(s)
- Shuang Fu
- a State Key Laboratory of Fine Chemicals , Dalian University of Technology , Dalian , China.,b Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education , Dalian Minzu University , Dalian , China
| | - Xiaodong Xu
- b Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education , Dalian Minzu University , Dalian , China
| | - Yu Ma
- b Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education , Dalian Minzu University , Dalian , China
| | - Shubiao Zhang
- b Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education , Dalian Minzu University , Dalian , China
| | - Shufen Zhang
- a State Key Laboratory of Fine Chemicals , Dalian University of Technology , Dalian , China
| |
Collapse
|
43
|
Mao B, Liu C, Zheng W, Li X, Ge R, Shen H, Guo X, Lian Q, Shen X, Li C. Cyclic cRGDfk peptide and Chlorin e6 functionalized silk fibroin nanoparticles for targeted drug delivery and photodynamic therapy. Biomaterials 2018; 161:306-320. [DOI: 10.1016/j.biomaterials.2018.01.045] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/03/2018] [Accepted: 01/27/2018] [Indexed: 12/13/2022]
|
44
|
Cheng H, Chi C, Shang W, Rengaowa S, Cui J, Ye J, Jiang S, Mao Y, Zeng C, Huo H, Chen L, Tian J. Precise integrin-targeting near-infrared imaging-guided surgical method increases surgical qualification of peritoneal carcinomatosis from gastric cancer in mice. Oncotarget 2018; 8:6258-6272. [PMID: 28009982 PMCID: PMC5351629 DOI: 10.18632/oncotarget.14058] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 12/12/2016] [Indexed: 12/23/2022] Open
Abstract
Peritoneal carcinomatosis from gastric cancer represents a common recurrent gastric cancer that seriously affects the survival, prognosis, and quality of life of patients at its advanced stage. In recent years, complete cytoreduction surgery in combination with hyperthermic intraperitoneal chemotherapy has been demonstrated to improve the survival and prognosis of patients with malignant tumors including peritoneal carcinomatosis from gastric cancer. Establishing viable methods of accurately assessing the tumor burden in patients with peritoneal carcinoma and correctly selecting suitable patients in order to improve cytoreduction surgical outcomes and reduce the risk of postoperative complications has become a challenge in the field of peritoneal carcinoma research. Here, we investigated peritoneal carcinomatosis from gastric cancer in a mouse model by using our self-developed surgical navigation system that combines optical molecular imaging with an integrin-targeting Arg-Gly-Asp-indocyanine green (RGD-ICG) molecular probe. The results showed that our diagnostic method could achieve a sensitivity and specificity of up to 93.93% and 100%, respectively, with a diagnostic index (DI) of 193.93% and diagnostic accuracy rate of 93.93%.Furthermore, the minimum tumor diameter measured during the surgery was 1.8 mm and the operative time was shortened by 3.26-fold when compared with the conventionally-treated control group. Therefore, our surgical navigation system that combines optical molecular imaging with an RGD-ICG molecular probe, could improve the diagnostic accuracy rate for peritoneal carcinomatosis from gastric cancer, shorten the operative time, and improve the quality of the cytoreduction surgery for peritoneal carcinomatosis from gastric cancer, thus providing a solid foundation for its future clinical development and application.
Collapse
Affiliation(s)
- Haidong Cheng
- Department of General Surgery, The Chinese PLA General Hospital, Beijing, 100853, China.,Department of General Surgery, The First Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, China.,Key Laboratory of Molecular Imaging of the Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Chongwei Chi
- Key Laboratory of Molecular Imaging of the Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wenting Shang
- Key Laboratory of Molecular Imaging of the Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Sha Rengaowa
- Department of Basic Medical Science, Inner Mongolia Medical University, Hohhot, 010059, China
| | - Jianxin Cui
- Department of General Surgery, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Jinzuo Ye
- Key Laboratory of Molecular Imaging of the Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Shixin Jiang
- Key Laboratory of Molecular Imaging of the Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yamin Mao
- Key Laboratory of Molecular Imaging of the Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Caoting Zeng
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Huiping Huo
- Department of Ultrasound, General Hospital of the People's Liberation Army, Beijing, 100853, China
| | - Lin Chen
- Department of General Surgery, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging of the Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
45
|
Anticancer Efficacy of Targeted Shikonin Liposomes Modified with RGD in Breast Cancer Cells. Molecules 2018; 23:molecules23020268. [PMID: 29382149 PMCID: PMC6017468 DOI: 10.3390/molecules23020268] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/04/2022] Open
Abstract
Shikonin (SHK) has been proven to have a good anti-tumor effect. However, poor water solubility and low bioavailability limit its wide application in clinical practice. In this study, to overcome these drawbacks, RGD-modified shikonin-loaded liposomes (RGD-SSLs-SHK) were successfully prepared. It exhibited excellent physicochemical characteristics including particle size, zeta potential, encapsulation efficiency, and delayed release time. Meanwhile, the targeting activity of the RGD-modified liposomes was demonstrated by flow cytometry and confocal microscopy in the αvβ3-positive MDA-MB-231 cells. Besides exhibiting greater cytotoxicity in vitro, compared with non-targeted shikonin-loaded liposomes (SSLs-SHK), RGD-SSLs-SHK could also evidently induce apoptosis by decreasing the expression of Bcl-2 and increasing the expression of Bax. It could also inhibit cell proliferation, migration, invasion, and adhesion by reducing the expression of MMP-9 and the level of NF-κB p65, but did not affect the expression of MMP-2 in the MDA-MB-231 cells. Therefore, these findings indicated that the strategy to use RGD-modified liposomes as carriers for targeted delivery of shikonin is a very promising approach to achieve breast cancer targeted therapy.
Collapse
|
46
|
Babu A, Amreddy N, Muralidharan R, Pathuri G, Gali H, Chen A, Zhao YD, Munshi A, Ramesh R. Chemodrug delivery using integrin-targeted PLGA-Chitosan nanoparticle for lung cancer therapy. Sci Rep 2017; 7:14674. [PMID: 29116098 PMCID: PMC5676784 DOI: 10.1038/s41598-017-15012-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/16/2017] [Indexed: 12/19/2022] Open
Abstract
In this study, we report the efficacy of RGD (arginine-glycine-aspartic acid) peptide-modified polylactic acid-co-glycolic acid (PLGA)-Chitosan nanoparticle (CSNP) for integrin αvβ3 receptor targeted paclitaxel (PTX) delivery in lung cancer cells and its impact on normal cells. RGD peptide-modified chitosan was synthesized and then coated onto PTX-PLGA nanoparticles prepared by emulsion-solvent evaporation. PTX-PLGA-CSNP-RGD displayed favorable physicochemical properties for a targeted drug delivery system. The PTX-PLGA-CSNP-RGD system showed increased uptake via integrin receptor mediated endocytosis, triggered enhanced apoptosis, and induced G2/M cell cycle arrest and more overall cytotoxicity than its non-targeted counterpart in cancer cells. PTX-PLGA-CSNP-RGD showed less toxicity in lung fibroblasts than in cancer cells, may be attributed to low drug sensitivity, nevertheless the study invited close attention to their transient overexpression of integrin αvβ3 and cautioned against corresponding uptake of toxic drugs, if any at all. Whereas, normal human bronchial epithelial (NHBE) cells with poor integrin αvβ3 expression showed negligible toxicity to PTX-PLGA-CSNP-RGD, at equivalent drug concentrations used in cancer cells. Further, the nanoparticle demonstrated its capacity in targeted delivery of Cisplatin (CDDP), a drug having physicochemical properties different to PTX. Taken together, our study demonstrates that PLGA-CSNP-RGD is a promising nanoplatform for integrin targeted chemotherapeutic delivery to lung cancer.
Collapse
Affiliation(s)
- Anish Babu
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.,Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Narsireddy Amreddy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.,Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Ranganayaki Muralidharan
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.,Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Gopal Pathuri
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.,Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Hariprasad Gali
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.,Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Allshine Chen
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.,Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Yan D Zhao
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.,Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Anupama Munshi
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.,Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Rajagopal Ramesh
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA. .,Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA. .,Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.
| |
Collapse
|
47
|
Alves Rico SR, Abbasi AZ, Ribeiro G, Ahmed T, Wu XY, de Oliveira Silva D. Diruthenium(ii,iii) metallodrugs of ibuprofen and naproxen encapsulated in intravenously injectable polymer-lipid nanoparticles exhibit enhanced activity against breast and prostate cancer cells. NANOSCALE 2017; 9:10701-10714. [PMID: 28678269 DOI: 10.1039/c7nr01582h] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
A unique class of diruthenium(ii,iii) metallodrugs containing non-steroidal anti-inflammatory drug (NSAID), Ru2(NSAID), have been reported to show anticancer activity in glioma models in vitro and in vivo. This work reports the encapsulation of the lead metallodrug of ibuprofen (HIbp), [Ru2(Ibp)4Cl] or RuIbp, and also of the new analogue of naproxen (HNpx), [Ru2(Npx)4Cl] or RuNpx, in novel intravenously (i.v.) injectable solid polymer-lipid nanoparticles (SPLNs). A rationally selected composition of lipids/polymers rendered nearly spherical Ru2(NSAID)-SPLNs with a mean size of 120 nm and zeta potential of about -20 mV. The Ru2(NSAID)-SPLNs are characterized by spectroscopic techniques and the composition in terms of ruthenium-drug species is analyzed by mass spectrometry. The metallodrug-loaded nanoparticles showed high drug loading (17-18%) with ∼100% drug loading efficiency, and good colloidal stability in serum at body temperature. Fluorescence-labeled SPLNs were taken up by the cancer cells in a time- and energy-dependent manner as analyzed by confocal microscopy and fluorescence spectrometry. The Ru2(NSAID)-SPLNs showed enhanced cytotoxicity (IC50 at 60-100 μmol L-1 ) in relation to the corresponding Ru2(NSAID) metallodrugs in breast (EMT6 and MDA-MB-231) and prostate (DU145) cancer cells in vitro. The cell viability of both metallodrug nanoformulations is also compared with those of the parent NSAIDs, HIbp and HNpx, and their corresponding NSAID-SPLNs. In vivo and ex vivo fluorescence imaging revealed good biodistribution and high tumor accumulation of fluorescence-labeled SPLNs following i.v. injection in an orthotopic breast tumor model. The enhanced anticancer activity of the metallodrug-loaded SPLNs in these cell lines can be associated with the advantages of the nanoformulations, assigned mainly to the stability of the colloidal nanoparticles suitable for i.v. injection and enhanced cellular uptake. The findings of this work encourage future in vivo efficacy studies to further exploit the potential of the novel Ru2(NSAID)-SPLN nanoformulations for clinical application.
Collapse
Affiliation(s)
- Samara R Alves Rico
- Laboratory for Synthetic and Structural Inorganic Chemistry - Bioinorganic and Metallodrugs, Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, B2 T, 05508-000, São Paulo, SP, Brazil.
| | | | | | | | | | | |
Collapse
|
48
|
Zhang T, Prasad P, Cai P, He C, Shan D, Rauth AM, Wu XY. Dual-targeted hybrid nanoparticles of synergistic drugs for treating lung metastases of triple negative breast cancer in mice. Acta Pharmacol Sin 2017; 38:835-847. [PMID: 28216624 PMCID: PMC5520182 DOI: 10.1038/aps.2016.166] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/26/2016] [Indexed: 12/21/2022] Open
Abstract
Lung metastasis is the major cause of death in patients with triple negative breast
cancer (TNBC), an aggressive subtype of breast cancer with no effective therapy at
present. It has been proposed that dual-targeted therapy, ie, targeting
chemotherapeutic agents to both tumor vasculature and cancer cells, may offer some
advantages. The present work was aimed to develop a dual-targeted synergistic drug
combination nanomedicine for the treatment of lung metastases of TNBC. Thus,
Arg-Gly-Asp peptide (RGD)-conjugated, doxorubicin (DOX) and mitomycin C (MMC)
co-loaded polymer-lipid hybrid nanoparticles (RGD-DMPLN) were prepared and
characterized. The synergism between DOX and MMC and the effect of RGD-DMPLN on cell
morphology and cell viability were evaluated in human MDA-MB-231 cells in
vitro. The optimal RGD density on nanoparticles (NPs) was identified based on
the biodistribution and tumor accumulation of the NPs in a murine lung metastatic
model of MDA-MB-231 cells. The microscopic distribution of RGD-conjugated NPs in lung
metastases was examined using confocal microscopy. The anticancer efficacy of
RGD-DMPLN was investigated in the lung metastatic model. A synergistic ratio of DOX
and MMC was found in the MDA-MB-231 human TNBC cells. RGD-DMPLN induced morphological
changes and enhanced cytotoxicity in vitro. NPs with a median RGD density
showed the highest accumulation in lung metastases by targeting both tumor
vasculature and cancer cells. Compared to free drugs, RGD-DMPLN exhibited
significantly low toxicity to the host, liver and heart. Compared to non-targeted
DMPLN or free drugs, administration of RGD-DMPLN (10 mg/kg, iv) resulted in a
4.7-fold and 31-fold reduction in the burden of lung metastases measured by
bioluminescence imaging, a 2.4-fold and 4.0-fold reduction in the lung metastasis
area index, and a 35% and 57% longer median survival time, respectively.
Dual-targeted RGD-DMPLN, with optimal RGD density, significantly inhibited the
progression of lung metastasis and extended host survival.
Collapse
|
49
|
Surface modification of lipid-based nanocarriers for cancer cell-specific drug targeting. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0329-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
50
|
Kuang Y, Zhang K, Cao Y, Chen X, Wang K, Liu M, Pei R. Hydrophobic IR-780 Dye Encapsulated in cRGD-Conjugated Solid Lipid Nanoparticles for NIR Imaging-Guided Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:12217-12226. [PMID: 28306236 DOI: 10.1021/acsami.6b16705] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
This is high demand to enhance the accumulation of near-infrared theranostic agents in the tumor region, which is favorable to the effective phototherapy. Compared with indocyanine green (a clinically applied dye), IR-780 iodide possesses higher and more stable fluorescence intensity and can be utilized as an imaging-guided PTT agent with laser irradiation. However, lipophilicity and short circulation time limit its applications in cancer imaging and therapy. Moreover, solid lipid nanoparticles (SLNs) conjugated with c(RGDyK) was designed as efficient carriers to improve the targeted delivery of IR-780 to the tumors. The multifunctional cRGD-IR-780 SLNs exhibited a desirable monodispersity, preferable stability and significant targeting to cell lines overexpressing αvβ3 integrin. Additionally, the in vitro assays such as cell viability and in vivo PTT treatment denoted that U87MG cells or U87MG transplantation tumors could be eradicated by applying cRGD-IR-780 SLNs under laser irradiation. Therefore, the resultant cRGD-IR-780 SLNs may serve as a promising NIR imaging-guided targeting PTT agent for cancer therapy.
Collapse
Affiliation(s)
- Ye Kuang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | - Kunchi Zhang
- Shanghai University of Medicine & Health Sciences , Shanghai 200120, China
| | - Yi Cao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | - Xing Chen
- Public Health of Guangxi Medical University , Nanning 530021, China
| | - Kewei Wang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | - Min Liu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| |
Collapse
|