1
|
Nikitovic D, Kukovyakina E, Berdiaki A, Tzanakakis A, Luss A, Vlaskina E, Yagolovich A, Tsatsakis A, Kuskov A. Enhancing Tumor Targeted Therapy: The Role of iRGD Peptide in Advanced Drug Delivery Systems. Cancers (Basel) 2024; 16:3768. [PMID: 39594723 PMCID: PMC11592346 DOI: 10.3390/cancers16223768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Chemotherapy remains the primary therapeutic approach in treating cancer. The tumor microenvironment (TME) is the complex network surrounding tumor cells, comprising various cell types, such as immune cells, fibroblasts, and endothelial cells, as well as ECM components, blood vessels, and signaling molecules. The often stiff and dense network of the TME interacts dynamically with tumor cells, influencing cancer growth, immune response, metastasis, and resistance to therapy. The effectiveness of the treatment of solid tumors is frequently reduced due to the poor penetration of the drug, which leads to attaining concentrations below the therapeutic levels at the site. Cell-penetrating peptides (CPPs) present a promising approach that improves the internalization of therapeutic agents. CPPs, which are short amino acid sequences, exhibit a high ability to pass cell membranes, enabling them to deliver drugs efficiently with minimal toxicity. Specifically, the iRGD peptide, a member of CPPs, is notable for its capacity to deeply penetrate tumor tissues by binding simultaneously integrins ανβ3/ανβ5 and neuropilin receptors. Indeed, ανβ3/ανβ5 integrins are characteristically expressed by tumor cells, which allows the iRGD peptide to home onto tumor cells. Notably, the respective dual-receptor targeting mechanism considerably increases the permeability of blood vessels in tumors, enabling an efficient delivery of co-administered drugs or nanoparticles into the tumor mass. Therefore, the iRGD peptide facilitates deeper drug penetration and improves the efficacy of co-administered therapies. Distinctively, we will focus on the iRGD mechanism of action, drug delivery systems and their application, and deliberate future perspectives in developing iRGD-conjugated therapeutics. In summary, this review discusses the potential of iRGD in overcoming barriers to drug delivery in cancer to maximize treatment efficiency while minimizing side effects.
Collapse
Affiliation(s)
- Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Ekaterina Kukovyakina
- Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (E.K.); (A.L.); (E.V.); (A.K.)
| | - Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Alexandros Tzanakakis
- School of Electrical and Computer Engineering, National Technical University of Athens, 15780 Athens, Greece;
| | - Anna Luss
- Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (E.K.); (A.L.); (E.V.); (A.K.)
| | - Elizaveta Vlaskina
- Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (E.K.); (A.L.); (E.V.); (A.K.)
| | - Anne Yagolovich
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Aristides Tsatsakis
- Forensic Medicine Department, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Andrey Kuskov
- Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia; (E.K.); (A.L.); (E.V.); (A.K.)
| |
Collapse
|
2
|
Hou X, Chen Q, Fang Y, Zhang L, Huang S, Xu M, Ren Y, Shi Z, Wei Y, Li L. iRGD-Guided Silica/Gold Nanoparticles for Efficient Tumor-Targeting and Enhancing Antitumor Efficacy Against Breast Cancer. Int J Nanomedicine 2024; 19:8237-8251. [PMID: 39157735 PMCID: PMC11329605 DOI: 10.2147/ijn.s474135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024] Open
Abstract
Background Breast cancer presents significant challenges due to the limited effectiveness of available treatments and the high likelihood of recurrence. iRGD possesses both RGD sequence and C-terminal sequence and has dual functions of targeting and membrane penetration. iRGD-modified nanocarriers can enhance drug targeting of tumor vascular endothelial cells and penetration of new microvessels, increasing drug concentration in tumor tissues. Methods The amidation reaction was carried out between SiO2/AuNCs and iRGD/PTX, yielding a conjugated drug delivery system (SiO2/AuNCs-iRGD/PTX, SAIP@NPs). The assessment encompassed the characterization of the morphology, particle size distribution, physicochemical properties, in vitro release profile, cytotoxicity, and cellular uptake of SAIP@NPs. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed using a small animal in vivo imaging system and a tumor-bearing nude mice model, respectively. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed utilizing a small animal in vivo imaging system and an in situ nude mice breast cancer xenograft model, respectively. Results The prepared SAIP@NPs exhibited decent stability and a certain slow-release effect in phosphate buffer (PBS, pH 7.4). In vitro studies had shown that, due to the dual functions of transmembrane and targeting of iRGD peptide, SAIP@NPs exhibited strong binding to integrin αvβ3, which was highly expressed on the membrane of MDA-MB-231 cells, improving the uptake capacity of tumor cells, inhibiting the rapid growth of tumor cells, and promoting tumor cell apoptosis. The results of animal experiments further proved that SAIP@NPs had longer residence time in tumor sites, stronger anti-tumor effect, and no obvious toxicity to major organs of experimental animals. Conclusion The engineered SAIP@NPs exhibited superior functionalities including efficient membrane permeability, precise tumor targeting, and imaging, thereby significantly augmenting the therapeutic efficacy against breast cancer with a favorable safety profile.
Collapse
Affiliation(s)
- Xuefeng Hou
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Qi Chen
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Ying Fang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Li Zhang
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Shuoheng Huang
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Minjie Xu
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Yaning Ren
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Zhansen Shi
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Yan Wei
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Lihua Li
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| |
Collapse
|
3
|
Thirumalai A, Girigoswami K, Pallavi P, Harini K, Gowtham P, Girigoswami A. Cancer therapy with iRGD as a tumor-penetrating peptide. Bull Cancer 2023; 110:1288-1300. [PMID: 37813754 DOI: 10.1016/j.bulcan.2023.08.009] [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: 07/03/2023] [Revised: 08/16/2023] [Accepted: 08/24/2023] [Indexed: 10/11/2023]
Abstract
One of the primary threats in tumor treatment revolves around the limited ability to penetrate tumor sites, leading to reduced therapeutic effectiveness, which remains a critical concern. Recently gaining importance are novel peptides, namely CRGDK/RGPD/EC (iRGD), that possess enhanced tumor-penetrating and inhibitory properties. These peptides specifically target and penetrate tumors by binding to αvβ integrins, namely αvβ3 and αvβ5, as well as NRP-1 receptors. Remarkably abundant on both the vasculature and tumor cell surfaces, these peptides show promising potential for improving tumor treatment outcomes. As a result, iRGD penetrated deep into the tumor tissues with biological products, contrast agents (imaging agents), antitumor drugs, and immune modulators after co-injecting them with peptides or chemically linked to peptides. The synthesis of iRGD peptides is a relatively straightforward process compared to the synthesis of other traditional peptides, and they significantly improved tumor tissue penetration inhibiting tumor metastasis effectively. Recent studies demonstrate the effectiveness of iRGD-driven dual-targeting chemotherapeutics on cancer cells, and the nanocarriers were modified with iRGD, serving as a favorable delivery strategy of payloads for deeper tumor regions. This review aims to provide an overview to emphasize the recent advancements and advantages of iRGD in treating and imaging various cancers.
Collapse
Affiliation(s)
- Anbazhagan Thirumalai
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Koyeli Girigoswami
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Pragya Pallavi
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Karthick Harini
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Pemula Gowtham
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Agnishwar Girigoswami
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India.
| |
Collapse
|
4
|
Xu M, Han X, Xiong H, Gao Y, Xu B, Zhu G, Li J. Cancer Nanomedicine: Emerging Strategies and Therapeutic Potentials. Molecules 2023; 28:5145. [PMID: 37446806 DOI: 10.3390/molecules28135145] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer continues to pose a severe threat to global health, making pursuing effective treatments more critical than ever. Traditional therapies, although pivotal in managing cancer, encounter considerable challenges, including drug resistance, poor drug solubility, and difficulties targeting tumors, specifically limiting their overall efficacy. Nanomedicine's application in cancer therapy signals a new epoch, distinguished by the improvement of the specificity, efficacy, and tolerability of cancer treatments. This review explores the mechanisms and advantages of nanoparticle-mediated drug delivery, highlighting passive and active targeting strategies. Furthermore, it explores the transformative potential of nanomedicine in tumor therapeutics, delving into its applications across various treatment modalities, including surgery, chemotherapy, immunotherapy, radiotherapy, photodynamic and photothermal therapy, gene therapy, as well as tumor diagnosis and imaging. Meanwhile, the outlook of nanomedicine in tumor therapeutics is discussed, emphasizing the need for addressing toxicity concerns, improving drug delivery strategies, enhancing carrier stability and controlled release, simplifying nano-design, and exploring novel manufacturing technologies. Overall, integrating nanomedicine in cancer treatment holds immense potential for revolutionizing cancer therapeutics and improving patient outcomes.
Collapse
Affiliation(s)
- Manman Xu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xinpu Han
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Hongtai Xiong
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yijie Gao
- Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Bowen Xu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guanghui Zhu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
5
|
Vyas K, Rathod M, Patel MM. Insight on nano drug delivery systems with targeted therapy in treatment of oral cancer. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 49:102662. [PMID: 36746272 DOI: 10.1016/j.nano.2023.102662] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Oral cancer is a type of cancer that develops in the mouth and is one of the deadliest malignancies in the world. Currently surgical, radiation therapy, and chemotherapy are most common treatments. Better treatment and early detection strategies are required. Chemotherapeutic drugs fail frequently due to toxicity and poor tumor targeting. There are high chances of failure of chemotherapeutic drugs due to toxicity. Active, passive, and immunity-targeting techniques are devised for tumor-specific activity. Nanotechnology-based drug delivery systems are the best available solution and important for precise targeting. Nanoparticles, liposomes, exosomes, and cyclodextrins are nano-based carriers for drug delivery. Nanotechnology is being used to develop new techniques such as intratumoral injections, microbubble mediated ultrasonic therapy, phototherapies, and site-specific delivery. This systematic review delves into the details of such targeted and nano-based drug delivery systems in order to improve patient health and survival rates in oral cancer.
Collapse
Affiliation(s)
- Kunj Vyas
- Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India
| | - Maharshsinh Rathod
- Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India
| | - Mayur M Patel
- Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India.
| |
Collapse
|
6
|
Liu C, Wu K, Li J, Mu X, Gao H, Xu X. Nanoparticle-mediated therapeutic management in cholangiocarcinoma drug targeting: Current progress and future prospects. Biomed Pharmacother 2023; 158:114135. [PMID: 36535198 DOI: 10.1016/j.biopha.2022.114135] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Patients with cholangiocarcinoma (CCA) often have an unfavorable prognosis because of its insidious nature, low resectability rate, and poor response to anticancer drugs and radiotherapy, which makes early detection and treatment difficult. At present, CCA has a five-year overall survival rate (OS) of only 5%, despite advances in therapies. New an increasing number of evidence suggests that nanoplatforms may play a crucial role in enhancing the pharmacological effects and in reducing both short- and long-term side effects of cancer treatment. This document reviews the advantages and shortcomings of nanoparticles such as liposomes, polymeric nanoparticle,inorganic nanoparticle, nano-metals and nano-alloys, carbon dots, nano-micelles, dendrimer, nano-capsule, bio-Nanomaterials in the diagnosis and treatment of CCA and discuss the current challenges in of nanoplatforms for CCA.
Collapse
Affiliation(s)
- Chunkang Liu
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jianyang Li
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xupeng Mu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Huan Gao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaohua Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
7
|
Ahmad A, Imran M, Sharma N. Precision Nanotoxicology in Drug Development: Current Trends and Challenges in Safety and Toxicity Implications of Customized Multifunctional Nanocarriers for Drug-Delivery Applications. Pharmaceutics 2022; 14:2463. [PMID: 36432653 PMCID: PMC9697541 DOI: 10.3390/pharmaceutics14112463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/06/2022] [Accepted: 11/13/2022] [Indexed: 11/17/2022] Open
Abstract
The dire need for the assessment of human and environmental endangerments of nanoparticulate material has motivated the formulation of novel scientific tools and techniques to detect, quantify, and characterize these nanomaterials. Several of these paradigms possess enormous possibilities for applications in many of the realms of nanotoxicology. Furthermore, in a large number of cases, the limited capabilities to assess the environmental and human toxicological outcomes of customized and tailored multifunctional nanoparticles used for drug delivery have hindered their full exploitation in preclinical and clinical settings. With the ever-compounded availability of nanoparticulate materials in commercialized settings, an ever-arising popular debate has been egressing on whether the social, human, and environmental costs associated with the risks of nanomaterials outweigh their profits. Here we briefly review the various health, pharmaceutical, and regulatory aspects of nanotoxicology of engineered multifunctional nanoparticles in vitro and in vivo. Several aspects and issues encountered during the safety and toxicity assessments of these drug-delivery nanocarriers have also been summarized. Furthermore, recent trends implicated in the nanotoxicological evaluations of nanoparticulate matter in vitro and in vivo have also been discussed. Due to the absence of robust and rigid regulatory guidelines, researchers currently frequently encounter a larger number of challenges in the toxicology assessment of nanocarriers, which have also been briefly discussed here. Nanotoxicology has an appreciable and significant part in the clinical translational development as well as commercialization potential of nanocarriers; hence these aspects have also been touched upon. Finally, a brief overview has been provided regarding some of the nanocarrier-based medicines that are currently undergoing clinical trials, and some of those which have recently been commercialized and are available for patients. It is expected that this review will instigate an appreciable interest in the research community working in the arena of pharmaceutical drug development and nanoformulation-based drug delivery.
Collapse
Affiliation(s)
- Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Mohammad Imran
- Therapeutics Research Group, Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane 4102, Australia
| | - Nisha Sharma
- Division of Nephrology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| |
Collapse
|
8
|
Alhaj-Suliman SO, Wafa EI, Salem AK. Engineering nanosystems to overcome barriers to cancer diagnosis and treatment. Adv Drug Deliv Rev 2022; 189:114482. [PMID: 35944587 DOI: 10.1016/j.addr.2022.114482] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 01/24/2023]
Abstract
Over the past two decades, multidisciplinary investigations into the development of nanoparticles for medical applications have continually increased. However, nanoparticles are still subject to biological barriers and biodistribution challenges, which limit their overall clinical potential. This has motivated the implementation of innovational modifications to a range of nanoparticle formulations designed for cancer imaging and/or cancer treatment to overcome specific barriers and shift the accumulation of payloads toward the diseased tissues. In recent years, novel technological and chemical approaches have been employed to modify or functionalize the surface of nanoparticles or manipulate the characteristics of nanoparticles. Combining these approaches with the identification of critical biomarkers provides new strategies for enhancing nanoparticle specificity for both cancer diagnostic and therapeutic applications. This review discusses the most recent advances in the design and engineering of nanoparticles as well as future directions for developing the next generation of nanomedicines.
Collapse
Affiliation(s)
- Suhaila O Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States; Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, United States.
| |
Collapse
|
9
|
Dual Targeting of Cancer Cells and MMPs with Self-Assembly Hybrid Nanoparticles for Combination Therapy in Combating Cancer. Pharmaceutics 2021; 13:pharmaceutics13121990. [PMID: 34959271 PMCID: PMC8707712 DOI: 10.3390/pharmaceutics13121990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/19/2021] [Indexed: 01/21/2023] Open
Abstract
The co-delivery of chemotherapeutic agents and immune modulators to their targets remains to be a great challenge for nanocarriers. Here, we developed a hybrid thermosensitive nanoparticle (TMNP) which could co-deliver paclitaxel-loaded transferrin (PTX@TF) and marimastat-loaded thermosensitive liposomes (MMST/LTSLs) for the dual targeting of cancer cells and the microenvironment. TMNPs could rapidly release the two payloads triggered by the hyperthermia treatment at the site of tumor. The released PTX@TF entered cancer cells via transferrin-receptor-mediated endocytosis and inhibited the survival of tumor cells. MMST was intelligently employed as an immunomodulator to improve immunotherapy by inhibiting matrix metalloproteinases to reduce chemokine degradation and recruit T cells. The TMNPs promoted the tumor infiltration of CD3+ T cells by 2-fold, including memory/effector CD8+ T cells (4.2-fold) and CD4+ (1.7-fold), but not regulatory T cells. Our in vivo anti-tumor experiment suggested that TMNPs possessed the highest tumor growth inhibitory rate (80.86%) compared with the control group. We demonstrated that the nanoplatform could effectively inhibit the growth of tumors and enhance T cell recruitment through the co-delivery of paclitaxel and marimastat, which could be a promising strategy for the combination of chemotherapy and immunotherapy for cancer treatment.
Collapse
|
10
|
Raveendran A, Poilil Surendran S, Ser J, Alam K, Cho H, Jeong YY. Heptamethine Cyanine Dye MHI-148-Mediated Drug Delivery System to Enhance the Anticancer Efficiency of Paclitaxel. Int J Nanomedicine 2021; 16:7169-7180. [PMID: 34707356 PMCID: PMC8545142 DOI: 10.2147/ijn.s325322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/20/2021] [Indexed: 12/19/2022] Open
Abstract
Introduction Paclitaxel (PTX) is a conventional chemotherapeutic drug that effectively treats various cancers. The cellular uptake and therapeutic potential of PTX are limited by its slow penetration and low solubility in water. The development of cancer chemotherapy methods is currently facing considerable challenges with respect to the delivery of the drugs, particularly in targeting the tumor site without exerting detrimental effects on the healthy surrounding cells. One possibility for improving the therapeutic potential is through the development of tumor-targeted delivery methods. Methods We successfully synthesized paclitaxel-MHI-148 conjugates (PTX-MHI) by coupling PTX with the tumor-targeting heptamethine cyanine dye MHI-148. Synthesis and purification were characterized using the absorbance spectrum and the results of time-of-flight mass spectrometry. Cellular uptake and cytotoxicity studies were conducted in vitro and in vivo. Results PTX-MHI accumulates in tumor cells but not in normal cells, as observed by in vitro near-infrared fluorescent (NIRF) imaging along with in vivo NIRF imaging and organ biodistribution studies. We observed that MHI-148-conjugated PTX shows greater efficiency in cancer cells than PTX alone, even in the absence of light treatment. PTX-MHI could also be used for specific drug delivery to intracellular compartments, such as the mitochondria and lysosomes of cancer cells, to improve the outcomes of tumor-targeting therapy. Conclusion The results indicated that PTX-MHI-mediated cancer therapy exerts an excellent inhibitory effect on colon carcinoma (HT-29) cell growth with low toxicity in normal fibroblasts (NIH3T3).
Collapse
Affiliation(s)
- Athira Raveendran
- Department of Materials Science & Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Suchithra Poilil Surendran
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Jinhui Ser
- Department of Materials Science & Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Khurshed Alam
- Department of Materials Science & Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hoonsung Cho
- Department of Materials Science & Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, 58128, Republic of Korea
| |
Collapse
|
11
|
Mishra K, Jain AK. Liposomes: An Emerging Approach for the Treatment of Cancer. Curr Pharm Des 2021; 27:2398-2414. [PMID: 33823772 DOI: 10.2174/1381612827666210406141449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/27/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Conventional drug delivery agents for a life-threatening disease, i.e., cancer, lack specificity towards cancer cells, producing a greater degree of side effects in the normal cells with a poor therapeutic index. These toxic side effects often limit dose escalation of anti-cancer drugs, leading to incomplete tumor suppression/ cancer eradication, early disease relapse, and ultimately, the development of drug resistance. Accordingly, targeting the tumor vasculatures is essential for the treatment of cancer. OBJECTIVE To search and describe a safer drug delivery carrier for the treatment of cancer with reduced systemic toxicities. METHOD Data were collected from Medline, PubMed, Google Scholar, Science Direct using the following keywords: 'liposomes', 'nanocarriers', 'targeted drug delivery', 'ligands', 'liposome for anti-cancerous drugs', 'treatment for cancer' and 'receptor targeting.' RESULTS Liposomes have provided a safe platform for the targeted delivery of encapsulated anti-cancer drugs for the treatment of cancer, which results in the reduction of the cytotoxic side effects of anti-cancer drugs on normal cells. CONCLUSION Liposomal targeting is a better emerging approach as an advanced drug delivery carrier with targeting ligands for anti-cancer agents.
Collapse
Affiliation(s)
- Keerti Mishra
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009 (C.G.), India
| | - Akhlesh K Jain
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009 (C.G.), India
| |
Collapse
|
12
|
Briolay T, Petithomme T, Fouet M, Nguyen-Pham N, Blanquart C, Boisgerault N. Delivery of cancer therapies by synthetic and bio-inspired nanovectors. Mol Cancer 2021; 20:55. [PMID: 33761944 PMCID: PMC7987750 DOI: 10.1186/s12943-021-01346-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As a complement to the clinical development of new anticancer molecules, innovations in therapeutic vectorization aim at solving issues related to tumor specificity and associated toxicities. Nanomedicine is a rapidly evolving field that offers various solutions to increase clinical efficacy and safety. MAIN: Here are presented the recent advances for different types of nanovectors of chemical and biological nature, to identify the best suited for translational research projects. These nanovectors include different types of chemically engineered nanoparticles that now come in many different flavors of 'smart' drug delivery systems. Alternatives with enhanced biocompatibility and a better adaptability to new types of therapeutic molecules are the cell-derived extracellular vesicles and micro-organism-derived oncolytic viruses, virus-like particles and bacterial minicells. In the first part of the review, we describe their main physical, chemical and biological properties and their potential for personalized modifications. The second part focuses on presenting the recent literature on the use of the different families of nanovectors to deliver anticancer molecules for chemotherapy, radiotherapy, nucleic acid-based therapy, modulation of the tumor microenvironment and immunotherapy. CONCLUSION This review will help the readers to better appreciate the complexity of available nanovectors and to identify the most fitting "type" for efficient and specific delivery of diverse anticancer therapies.
Collapse
Affiliation(s)
- Tina Briolay
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | - Morgane Fouet
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | | | | |
Collapse
|
13
|
Lotfalian S, Nematollahzadeh A, Ghasemi S. Hierarchically structured protein-based hollow-nanospheres for drug delivery. REACT FUNCT POLYM 2021. [DOI: 10.1016/j.reactfunctpolym.2021.104821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
14
|
Crosslinked casein micelles bound paclitaxel as enzyme activated intracellular drug delivery systems for cancer therapy. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2020.110237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
15
|
Perrelli A, Fatehbasharzad P, Benedetti V, Ferraris C, Fontanella M, De Luca E, Moglianetti M, Battaglia L, Retta SF. Towards precision nanomedicine for cerebrovascular diseases with emphasis on Cerebral Cavernous Malformation (CCM). Expert Opin Drug Deliv 2021; 18:849-876. [PMID: 33406376 DOI: 10.1080/17425247.2021.1873273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Cerebrovascular diseases encompass various disorders of the brain vasculature, such as ischemic/hemorrhagic strokes, aneurysms, and vascular malformations, also affecting the central nervous system leading to a large variety of transient or permanent neurological disorders. They represent major causes of mortality and long-term disability worldwide, and some of them can be inherited, including Cerebral Cavernous Malformation (CCM), an autosomal dominant cerebrovascular disease linked to mutations in CCM1/KRIT1, CCM2, or CCM3/PDCD10 genes.Areas covered: Besides marked clinical and etiological heterogeneity, some commonalities are emerging among distinct cerebrovascular diseases, including key pathogenetic roles of oxidative stress and inflammation, which are increasingly recognized as major disease hallmarks and therapeutic targets. This review provides a comprehensive overview of the different clinical features and common pathogenetic determinants of cerebrovascular diseases, highlighting major challenges, including the pressing need for new diagnostic and therapeutic strategies, and focusing on emerging innovative features and promising benefits of nanomedicine strategies for early detection and targeted treatment of such diseases.Expert opinion: Specifically, we describe and discuss the multiple physico-chemical features and unique biological advantages of nanosystems, including nanodiagnostics, nanotherapeutics, and nanotheranostics, that may help improving diagnosis and treatment of cerebrovascular diseases and neurological comorbidities, with an emphasis on CCM disease.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Parisa Fatehbasharzad
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Valerio Benedetti
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Chiara Ferraris
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, Torino, Italy
| | - Marco Fontanella
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Elisa De Luca
- Nanobiointeractions & Nanodiagnostics, Center for Biomolecular Nanotechnologies, Arnesano, Lecce, Italy.,Institute for Microelectronics and Microsystems (IMM), CNR, Lecce, Italy
| | - Mauro Moglianetti
- Nanobiointeractions & Nanodiagnostics, Center for Biomolecular Nanotechnologies, Arnesano, Lecce, Italy.,Istituto Italiano Di Tecnologia, Nanobiointeractions & Nanodiagnostics, Genova, Italy
| | - Luigi Battaglia
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, Torino, Italy
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| |
Collapse
|
16
|
Kang S, Lee S, Park S. iRGD Peptide as a Tumor-Penetrating Enhancer for Tumor-Targeted Drug Delivery. Polymers (Basel) 2020; 12:E1906. [PMID: 32847045 PMCID: PMC7563641 DOI: 10.3390/polym12091906] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
The unique structure and physiology of a tumor microenvironment impede intra-tumoral penetration of chemotherapeutic agents. A novel iRGD peptide that exploits the tumor microenvironment can activate integrin-dependent binding to tumor vasculatures and neuropilin-1 (NRP-1)-dependent transport to tumor tissues. Recent studies have focused on its dual-targeting ability to achieve enhanced penetration of chemotherapeutics for the efficient eradication of cancer cells. Both the covalent conjugation and the co-administration of iRGD with chemotherapeutic agents and engineered delivery vehicles have been explored. Interestingly, the iRGD-mediated drug delivery also enhances penetration through the blood-brain barrier (BBB). Recent studies have shown its synergistic effect with BBB disruptive techniques. The efficacy of immunotherapy involving immune checkpoint blockades has also been amplified by using iRGD as a targeting moiety. In this review, we presented the recent advances in iRGD technology, focusing on cancer treatment modalities, including the current clinical trials using iRGD. The iRGD-mediated nano-carrier system could serve as a promising strategy in drug delivery to the deeper tumor regions, and be combined with various therapeutic interventions due to its novel targeting ability.
Collapse
Affiliation(s)
| | | | - Soyeun Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Korea; (S.K.); (S.L.)
| |
Collapse
|
17
|
Chen X, Hu C, Zhang Y, Hao W, He X, Li Q, Huang Y, Huang Y, Chen Y. Anticancer Activity and Mechanism of Action of kla-TAT Peptide. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10019-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Beltrán-Gracia E, López-Camacho A, Higuera-Ciapara I, Velázquez-Fernández JB, Vallejo-Cardona AA. Nanomedicine review: clinical developments in liposomal applications. Cancer Nanotechnol 2019. [DOI: 10.1186/s12645-019-0055-y] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract
Background
In recent years, disease treatment has evolved strategies that require increase in pharmaceutical agent’s efficacy and selectivity while decreasing their toxicity in normal tissues. These requirements have led to the development of nanoscale liposome systems for drug release. This review focuses on lipid features, pharmacological properties of liposomal formulations and the clinical studies of their application.
Main body
Several lipids are available, but their properties could affect pharmacological or clinical efficiency of drug formulations. Many liposomal formulations have been developed and are currently on the market. Proper selection of lipid is essential for the pharmacological effect to be improved. Most of the formulations use mainly zwitterionic, cationic or anionic lipids, PEG and/or cholesterol, which have different effects on stability, pharmacokinetics and delivery of the drug formulation. Clinical trials have shown that liposomes are pharmacologically and pharmacokinetically more efficient than drug-alone formulations in treating acute myeloid leukemia, hepatitis A, pain management, ovary, gastric breast and lung cancer, among others.
Conclusion
Liposomal formulations are less toxic than drugs alone and have better pharmacological parameters. Although they seem to be the first choice for drug delivery systems for various diseases, further research about dosage regimen regarding dose and time needs to be carried out.
Collapse
|
19
|
Li Z, Du X, Cui X, Wang Z. Ultrasonic-assisted fabrication and release kinetics of two model redox-responsive magnetic microcapsules for hydrophobic drug delivery. ULTRASONICS SONOCHEMISTRY 2019; 57:223-232. [PMID: 31078396 DOI: 10.1016/j.ultsonch.2019.04.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/12/2019] [Accepted: 04/26/2019] [Indexed: 05/27/2023]
Abstract
The smart biocompatible carriers have an advantage in the high-efficiency delivery and stimuli-responsive release of drugs. This study describes two model magnetic microcapsules (MMC) fabricated by sonicating the hydrophobic drug-loaded oil phase in an albumin aqueous solution, where magnetic nanoparticles are either encapsulated into the core or embedded onto the albumin shell. The as-prepared MMC with magnetic shell (MS) or with magnetic core (MC) shows an appropriate dispersibility with a well-defined spherical morphology in water, an excellent magnetism-mediated shifting ability for targeted drug delivery, and a good biocompatibility for high-level cell viability. Moreover, both the two microcapsules also show a high efficiency to trap the hydrophobic drugs, where the embedding ratios are 87.41% for MMC-MS and 95.31% for MMC-MC, respectively. Meanwhile in current study, the release kinetics and mechanism reveal that the sulfhydryl-crosslinked shell structure endows the MMC with a redox-responsive behavior to release the contents for controlled drug release, and the release rate or the release amount can be adjusted by changing the dosage of reducing agent. Therefore, the MMC have great potential as a smart carrier of hydrophobic drugs for enhancing the therapeutic efficiency.
Collapse
Affiliation(s)
- Zhanfeng Li
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, 266071 Qingdao, China; College of Chemistry, Jilin University, 130012 Changchun, China
| | - Xiaoyu Du
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, 266071 Qingdao, China
| | - Xuejun Cui
- College of Chemistry, Jilin University, 130012 Changchun, China.
| | - Zonghua Wang
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, 266071 Qingdao, China.
| |
Collapse
|
20
|
iRGD: A Promising Peptide for Cancer Imaging and a Potential Therapeutic Agent for Various Cancers. JOURNAL OF ONCOLOGY 2019; 2019:9367845. [PMID: 31346334 PMCID: PMC6617877 DOI: 10.1155/2019/9367845] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022]
Abstract
Poor penetration into the tumor parenchyma and the reduced therapeutic efficacy of anticancer drugs and other medications are the major problems in tumor treatment. A new tumor-homing and penetrating peptide, iRGD (CRGDK/RGPD/EC), can be effectively used to combine and deliver imaging agents or anticancer drugs into tumors. The different “vascular zip codes” expressed in different tissues can serve as targets for docking-based (synaptic) delivery of diagnostic and therapeutic molecules. αv-Integrins are abundantly expressed in the tumor vasculature, where they are recognized by peptides containing the RGD integrin recognition motif. The iRGD peptide follows a multistep tumor-targeting process: First, it is proteolytically cleaved to generate the CRGDK fragment by binding to the surface of cells expressing αv integrins (αvβ3 and αvβ5). Then, the fragment binds to neuropilin-1 and penetrates the tumor parenchyma more deeply. Compared with conventional RGD peptides, the affinity of iRGD for αv integrins is in the mid to low nanomolar range, and the CRGDK fragment has a stronger affinity for neuropilin-1 than that for αv integrins because of the C-terminal exposure of a conditional C-end Rule (CendR) motif (R/KXXR/K), whose receptor proved to be neuropilin-1. Consequently, these advantages facilitate the transfer of CRGDK fragments from integrins to neuropilin-1 and consequently deeper penetration into the tumor. Due to its specific binding and strong affinity, the iRGD peptide can deliver imaging agents and anticancer drugs into tumors effectively and deeply, which is useful in detecting the tumor, blocking tumor growth, and inhibiting tumor metastasis. This review aims to focus on the role of iRGD in the imaging and treatment of various cancers.
Collapse
|
21
|
Xiong K, Wu J, Liu Y, Wu N, Ruan J. Drug Carrier-Oriented Polygeline for Preparing Novel Polygeline-Bound Paclitaxel Nanoparticles. J Pharm Sci 2019; 108:2012-2021. [PMID: 30639741 DOI: 10.1016/j.xphs.2019.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 12/15/2018] [Accepted: 01/04/2019] [Indexed: 10/27/2022]
Abstract
Polygeline is a highly promising drug carrier-oriented material for important applications in pharmacy field due to its low-cost and unique properties similar to albumin. In this study, polygeline-bound paclitaxel nanoparticles (Npb-PTXS) were fabricated through a combination of low-pressure emulsification and high-pressure homogenization. The effects of a series of production parameters on mean particle size, particle size distribution and drug loading of Npb-PTXS were systematically evaluated. The characteristics of Npb-PTXS, such as surface morphology, physical status of paclitaxel (PTX) in Npb-PTXS, redispersibility of Npb-PTXS in purified water and bioavailability in vivo were also investigated. It is revealed that the optimal preparation conditions included an aqueous phase pH value of about 6.5, protein mass concentration of 0.33%, with mass ratio of PTX to protein of 30%, high pressure of 1200 bar, high-pressure passes of 25 times and low-pressure emulsifying passes of 20 times. Obtained Npb-PTXS shows good resolubility compared to commercially available Abraxane®, containing round or oval shaped particles with mean particle size of around 188.3 nm, polydispersity index of 0.163 and zeta potential of -31.1 mV. PTX in Npb-PTX is amorphous, and its content is approximately 12.04%. Encapsulation efficiency of Npb-PTXS reaches 81.2%. Moreover, in vivo pharmacokinetic studies showed that the intravenous relative bioavailability of Npb-PTXS to Abraxane was 83.89%.
Collapse
Affiliation(s)
- Kaibin Xiong
- School of Life Science, Wuchang University of Technology, Wuhan 430223, PR China
| | - Jianyang Wu
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Jiujiang Research, Institute and Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Xiamen 361005, PR China.
| | - Yang Liu
- School of Life Science, Wuchang University of Technology, Wuhan 430223, PR China
| | - Na Wu
- School of Life Science, Wuchang University of Technology, Wuhan 430223, PR China
| | - Jinlan Ruan
- School of Life Science, Wuchang University of Technology, Wuhan 430223, PR China.
| |
Collapse
|
22
|
Rathod V, Tripathi R, Joshi P, Jha PK, Bahadur P, Tiwari S. Paclitaxel Encapsulation into Dual-Functionalized Multi-Walled Carbon Nanotubes. AAPS PharmSciTech 2019; 20:51. [PMID: 30617845 DOI: 10.1208/s12249-018-1218-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/08/2018] [Indexed: 01/31/2023] Open
Abstract
This work reports the synthesis of multi-walled carbon nanotubes (CNTs) from xylene/ferrocene using catalytic chemical vapor deposition technique. Following characterization using transmission electron microscopy (TEM), energy-dispersive X-ray (EDX), and Raman spectroscopy, CNT surface was dual-functionalized using ethylenediamine and phenylboronic acid groups. Average diameter of CNTs was calculated to be 16.5 nm. EDX spectra confirmed the existence of carbonaceous deposits on the tube's surface. Scattered electron diffraction and X-ray peak broadening calculations showed consistent inter-planer distance of the grown CNTs. Chemical functionalization, confirmed from FT-IR and Raman spectra, showed an enhanced dispersibility of CNTs in water. We describe the changes in the first- and second-order regions of the Raman spectra following the encapsulation of an anti-cancer drug, paclitaxel (PLX), into the free volume of functionalized CNTs. High PLX loading, achieved through its non-covalent π-π stacking within the CNT interior, is confirmed through the blue-shifted, softened G band in the Raman spectrum. While not addressed here, we will exploit this dual functionalization tactic to elaborate the relative role of attached moieties in the affinity interaction of CNTs with extra-cellular sialic acid, a biological target showing metastatic stage-dependent over-expression in colon cancer cells.
Collapse
|
23
|
Lipid-bilayer-coated nanogels allow for sustained release and enhanced internalization. Int J Pharm 2018; 551:8-13. [DOI: 10.1016/j.ijpharm.2018.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/04/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
|
24
|
Golombek SK, May JN, Theek B, Appold L, Drude N, Kiessling F, Lammers T. Tumor targeting via EPR: Strategies to enhance patient responses. Adv Drug Deliv Rev 2018; 130:17-38. [PMID: 30009886 PMCID: PMC6130746 DOI: 10.1016/j.addr.2018.07.007] [Citation(s) in RCA: 783] [Impact Index Per Article: 130.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
Abstract
The tumor accumulation of nanomedicines relies on the enhanced permeability and retention (EPR) effect. In the last 5-10 years, it has been increasingly recognized that there is a large inter- and intra-individual heterogeneity in EPR-mediated tumor targeting, explaining the heterogeneous outcomes of clinical trials in which nanomedicine formulations have been evaluated. To address this heterogeneity, as in other areas of oncology drug development, we have to move away from a one-size-fits-all tumor targeting approach, towards methods that can be employed to individualize and improve nanomedicine treatments. To this end, efforts have to be invested in better understanding the nature, the complexity and the heterogeneity of the EPR effect, and in establishing systems and strategies to enhance, combine, bypass and image EPR-based tumor targeting. In the present manuscript, we summarize key studies in which these strategies are explored, and we discuss how these approaches can be employed to enhance patient responses.
Collapse
Affiliation(s)
- Susanne K Golombek
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Jan-Niklas May
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Benjamin Theek
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Lia Appold
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Natascha Drude
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Department of Nuclear Medicine, RWTH Aachen University Clinic, Aachen, Germany
| | - Fabian Kiessling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Department of Pharmaceutics, Utrecht University, Utrecht, the Netherlands; Department of Targeted Therapeutics, University of Twente, Enschede, the Netherlands.
| |
Collapse
|
25
|
Lv Y, Xu C, Zhao X, Lin C, Yang X, Xin X, Zhang L, Qin C, Han X, Yang L, He W, Yin L. Nanoplatform Assembled from a CD44-Targeted Prodrug and Smart Liposomes for Dual Targeting of Tumor Microenvironment and Cancer Cells. ACS NANO 2018; 12:1519-1536. [PMID: 29350904 DOI: 10.1021/acsnano.7b08051] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The tumor microenvironment (TME) plays a critical role in tumor initiation, progression, invasion, and metastasis. Therefore, a therapy that combines chemotherapeutic drugs with a TME modulator could be a promising route for cancer treatment. This paper reports a nanoplatform self-assembled from a hyaluronic acid (HA)-paclitaxel (PTX) (HA-PTX) prodrug and marimastat (MATT)-loaded thermosensitive liposomes (LTSLs) (MATT-LTSLs) for the dual targeting of the TME and cancer cells. Interestingly, the prodrug HA-PTX can self-assemble on both positively and negatively charged liposomes, forming hybrid nanoparticles (HNPs, 100 nm). Triggered by mild hyperthermia, HA-PTX/MATT-LTSLs HNPs rapidly release their payloads into the extracellular environment, and the released HA-PTX quickly enters 4T1 cells through a CD44-HA affinity. The HNPs possess promoted tumor accumulation (1.6-fold), exhibit deep tumor penetration, and significantly inhibit the tumor growth (10-fold), metastasis (100%), and angiogenesis (10-fold). Importantly, by targeting the TME and maintaining its integrity via inhibiting the expression and activity of matrix metalloproteinases (>5-fold), blocking the fibroblast activation by downregulating the TGF-β1 expression (5-fold) and suppressing the degradation of extracellular matrix, the HNPs allow for significant metastasis inhibition. Overall, these findings indicate that a prodrug of an HA-hydrophobic-active compound and liposomes can be self-assembled into a smart nanoplatform for the dual targeting of the TME and tumor cells and efficient combined treatment; additionally, the co-delivery of MATT and HA-PTX with the HNPs is a promising approach for the treatment of metastatic cancer. This study creates opportunities for fabricating multifunctional nanodevices and offers an efficient strategy for disease therapy.
Collapse
Affiliation(s)
- Yaqi Lv
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Chaoran Xu
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Xiangmei Zhao
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Chenshi Lin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Xin Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Xiaofei Xin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Li Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Chao Qin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Xiaopeng Han
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Lei Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Lifang Yin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| |
Collapse
|
26
|
Hu C, Ma N, Li F, Fang Y, Liu Y, Zhao L, Qiao S, Li X, Jiang X, Li T, Shen F, Huang Y, Luo Q, Liu J. Cucurbit[8]uril-Based Giant Supramolecular Vesicles: Highly Stable, Versatile Carriers for Photoresponsive and Targeted Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:4603-4613. [PMID: 29333854 DOI: 10.1021/acsami.8b00297] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Highly stable giant supramolecular vesicles were constructed by hierarchical self-assembly of cucurbit[8]uril (CB[8])-based supra-amphiphiles for photoresponsive and targeted intracellular drug delivery. These smart vesicles can encapsulate the model drugs with high loading efficiencies and then release them by manipulating photoswitchable CB[8] heteroternary complexation to regulate the formation and dissociation of supra-amphiphiles that cause dramatic morphological changes of the assemblies to achieve remote optically controlled drug delivery. More importantly, the confocal microscopy analysis, cellular uptake experiment, and cell viability assay have shown that the giant vesicles are able to maintain the structural integrity and stability within actual cellular environments and exhibit obvious advantages for intracellular drug delivery such as low toxicity, easy surface modification for tumor-targeting selectivity, and rapid internalization into different human cancer cell lines. A synergistic mechanism that integrates multiple pathways including energy-dependent endocytosis, macropinocytosis, cholesterol-dependent endocytosis, and microtubule-related endocytosis was determined to facilitate the internalization process. Moreover, cytotoxicity experiments and flow cytometric analysis have demonstrated that the doxorubicin hydrochloride-loaded vesicles exhibited a significant therapeutic effect for tumor cells upon UV light irradiation, which makes the photoresponsive system more promising for potential applications in pharmaceutically relevant fields.
Collapse
Affiliation(s)
- Cuihua Hu
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Ningning Ma
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Fei Li
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Yu Fang
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Yao Liu
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Linlu Zhao
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Shanpeng Qiao
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Xiumei Li
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Xiaojia Jiang
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Tiezhu Li
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Fangzhong Shen
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Yibing Huang
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Quan Luo
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| | - Junqiu Liu
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, and ‡Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University , 2699 Qianjin Road, Changchun 130012, China
| |
Collapse
|
27
|
Jin Z, Wu K, Hou J, Yu K, Shen Y, Guo S. A PTX/nitinol stent combination with temperature-responsive phase-change 1-hexadecanol for magnetocaloric drug delivery: Magnetocaloric drug release and esophagus tissue penetration. Biomaterials 2018; 153:49-58. [DOI: 10.1016/j.biomaterials.2017.10.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 01/12/2023]
|
28
|
Naqvi S, Mohiyuddin S, Gopinath P. Niclosamide loaded biodegradable chitosan nanocargoes: an in vitro study for potential application in cancer therapy. ROYAL SOCIETY OPEN SCIENCE 2017; 4:170611. [PMID: 29291056 PMCID: PMC5717630 DOI: 10.1098/rsos.170611] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/12/2017] [Indexed: 05/08/2023]
Abstract
Chitosan nanoparticles can advance the pharmacological and therapeutic properties of chemotherapeutic agents by controlling release rates and targeted delivery process, which eliminates the limitations of conventional anti-cancer therapies and they are also safe as well as cost-effective. The aim of present study is to explore the anti-tumour effect of niclosamide in lung and breast cancer cell lines using biocompatible and biodegradable carrier where nanoparticles loaded with hydrophobic drug (niclosamide) were synthesized, characterized and applied as a stable anti-cancer agent. Niclosamide loaded chitosan nanoparticles (Nic-Chi Np's) of size approximately 100-120 nm in diameter containing hydrophobic anti-cancer drug, i.e. niclosamide, were prepared. Physico-chemical characterization confirms that the prepared nanoparticles are spherical, monodispersed and stable in aqueous systems. The therapeutic efficacy of Nic-Chi Np's was evaluated against breast cancer cell line (MCF-7) and human lung cancer cell line (A549). MTT assay reveals the cell viability of the prepared Nic-Chi Np's against A549 and MCF-7 cells and obtained an IC50 value of 8.75 µM and 7.5 µM, respectively. Acridine orange/ethidium bromide dual staining results verified the loss of the majority of the cells by apoptosis. Flow cytometer analysis quantified the generation of intracellular reactive oxygen species (ROS) and signified that exposure to a higher concentration (2 × IC50) of Nic-Chi Np's resulted in elevated ROS generation. Notably, Nic-Chi Np treatment showed more apoptosis and cell death in MCF-7 as compared to A549. Further, the remarkable induction of apoptosis by Nic-Chi Np's was confirmed by semi-quantitative reverse transcription polymerase chain reaction, scanning electron microscopy and cell-cycle analysis. Thus, Nic-Chi Np's may have a great potential even at low concentration for anti-cancer therapy and may replace or substitute more toxic anti-mitotic drugs in the near future.
Collapse
Affiliation(s)
- Saba Naqvi
- Nanobiotechnology Laboratory, Centre of Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Shanid Mohiyuddin
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - P. Gopinath
- Nanobiotechnology Laboratory, Centre of Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
- Author for correspondence: P. Gopinath e-mail: ;
| |
Collapse
|
29
|
Advances in structural design of lipid-based nanoparticle carriers for delivery of macromolecular drugs, phytochemicals and anti-tumor agents. Adv Colloid Interface Sci 2017; 249:331-345. [PMID: 28477868 DOI: 10.1016/j.cis.2017.04.006] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/13/2017] [Accepted: 04/17/2017] [Indexed: 12/19/2022]
Abstract
The present work highlights recent achievements in development of nanostructured dispersions and biocolloids for drug delivery applications. We emphasize the key role of biological small-angle X-ray scattering (BioSAXS) investigations for the nanomedicine design. A focus is given on controlled encapsulation of small molecular weight phytochemical drugs in lipid-based nanocarriers as well as on encapsulation of macromolecular siRNA, plasmid DNA, peptide and protein pharmaceuticals in nanostructured nanoparticles that may provide efficient intracellular delivery and triggered drug release. Selected examples of utilisation of the BioSAXS method for characterization of various types of liquid crystalline nanoorganizations (liposome, spongosome, cubosome, hexosome, and nanostructured lipid carriers) are discussed in view of the successful encapsulation and protection of phytochemicals and therapeutic biomolecules in the hydrophobic or the hydrophilic compartments of the nanocarriers. We conclude that the structural design of the nanoparticulate carriers is of crucial importance for the therapeutic outcome and the triggered drug release from biocolloids.
Collapse
|
30
|
Liu X, Jiang J, Ji Y, Lu J, Chan R, Meng H. Targeted drug delivery using iRGD peptide for solid cancer treatment. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2017; 2:370-379. [PMID: 30498580 PMCID: PMC6258069 DOI: 10.1039/c7me00050b] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Many solid tumor types, such as pancreatic cancer, have a generally poor prognosis, in part because the delivery of therapeutic regimen is prohibited by pathological abnormalities that block access to tumor vasculature, leading to poor bioavailability. Recent development of tumor penetrating iRGD peptide that is covalently conjugated on nanocarriers' surface or co-administered with nanocarriers becomes a popular approach for tumor targeting. More importantly, scientists have unlocked an important tumor transcytosis mechanism by which drug carrying nanoparticles directly access solid tumors (without the need of leaky vasculature), thereby allowing systemically injected nanocarriers more abundantly distribute at tumor site with improved efficacy. In this focused review, we summarized the design and implementation strategy for iRGD-mediated tumor targeting. This includes the working principle of such peptide and discussion on patient-specific iRGD effect in vivo, commensurate with the level of key biomarker (i.e. neuropilin-1) expression on tumor vasculature. This highlights the necessity to contemplate the use of a personalized approach when iRGD technology is used in clinic.
Collapse
Affiliation(s)
- Xiangsheng Liu
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Jinhong Jiang
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Ying Ji
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Jianqin Lu
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Ryan Chan
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Huan Meng
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
31
|
Schwarzl R, Du F, Haag R, Netz RR. General method for the quantification of drug loading and release kinetics of nanocarriers. Eur J Pharm Biopharm 2017; 116:131-137. [DOI: 10.1016/j.ejpb.2016.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/20/2016] [Accepted: 12/20/2016] [Indexed: 10/20/2022]
|
32
|
Zhang L, Yang X, Lv Y, Xin X, Qin C, Han X, Yang L, He W, Yin L. Cytosolic co-delivery of miRNA-34a and docetaxel with core-shell nanocarriers via caveolae-mediated pathway for the treatment of metastatic breast cancer. Sci Rep 2017; 7:46186. [PMID: 28383524 PMCID: PMC5382875 DOI: 10.1038/srep46186] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/10/2017] [Indexed: 01/19/2023] Open
Abstract
Co-delivery of microRNAs and chemotherapeutic drugs into tumor cells is an attractive strategy for synergetic breast cancer therapy due to their complementary mechanisms. In this work, a core-shell nanocarrier coated by cationic albumin was developed to simultaneously deliver miRNA-34a and docetaxel (DTX) into breast cancer cells for improved therapeutic effect. The co-delivery nanocarriers showed a spherical morphology with an average particle size of 183.9 nm, and they efficiently protected miRNA-34a from degradation by RNase and serum. Importantly, the nanocarriers entered the cytosol via a caveolae-mediated pathway without entrapment in endosomes/lysosomes, thus improving the utilization of the cargo. In vitro, the co-delivery nanocarriers suppressed the expression of anti-apoptosis gene Bcl-2 at both transcription and protein levels, inhibited tumor cell migration and efficiently induced cell apoptosis and cytotoxicity. In vivo, the co-delivery nanocarriers prolonged the blood circulation of DTX, enhanced tumor accumulation of the cargo and significantly inhibited tumor growth and metastasis in 4T1-tumor bearing mice models. Taken together, the present nanocarrier co-loading with DTX and miRNA-34a is a new nanoplatform for the combination of insoluble drugs and gene/protein drugs and provides a promising strategy for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Xin Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yaqi Lv
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Xiaofei Xin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Chao Qin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Xiaopeng Han
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Lei Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Lifang Yin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China.,Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, P.R. China
| |
Collapse
|
33
|
Self-assembly of glutamic acid linked paclitaxel dimers into nanoparticles for chemotherapy. Bioorg Med Chem Lett 2017; 27:2493-2496. [PMID: 28404373 DOI: 10.1016/j.bmcl.2017.03.101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/21/2017] [Accepted: 03/31/2017] [Indexed: 12/11/2022]
Abstract
In this work, a glutamic acid linked paclitaxel (PTX) dimer (Glu-PTX2) with high PTX content of 88.9wt% was designed and synthesized. Glu-PTX2 could self-assemble into nanoparticles (Glu-PTX2 NPs) in aqueous solution to increase the water solubility of PTX. Glu-PTX2 NPs were characterized by electron microscopy and dynamic light scattering, exhibiting spherical morphology and favorable structural stability in aqueous media. Glu-PTX2 NPs could be internalized by cancer cells as revealed by confocal laser scanning microscopy and exert potent cytotoxicity. It is envisaged that Glu-PTX2 NPs would be an alternative formulation for PTX, and such amino acid linked drug dimers could also be applied to other therapeutic agents.
Collapse
|
34
|
He W, Wang Y, Lv Y, Xiao Q, Ye L, Cai B, Qin C, Han X, Cai T, Yin L. Denatured protein stabilized drug nanoparticles: tunable drug state and penetration across the intestinal barrier. J Mater Chem B 2017; 5:1081-1097. [PMID: 32263886 DOI: 10.1039/c6tb02577c] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanosuspensions of drugs are nanosized colloidal dispersions of pure particles. In contrast to conventional nanoparticles, the particles in nanosuspensions feature 100% drug loading. Stiripentol (STP) is an effective drug for severe myoclonic epilepsy of infancy (SMEI); however, because of its low water solubility, high oral doses of STP, up to 50 mg per kg per day in two or three divided doses, must be administered to patients, compromising therapy outcomes. Here, we report STP nanosuspensions (STP-Ns) stabilized with denatured soybean protein isolate (SPI) as a stabilizer to promote the absorption of STP and thus improve therapeutic outcomes. STP-Ns with a drug loading of up to 50% (w/w) and a diameter of 150 nm were successfully prepared. Importantly, in the presence of denatured SPI as a stabilizer, the drug state in the nanosuspensions was tunable by drug loading: low drug loading resulted in the formation of amorphous drug nanoparticles while high drug loading greater than 3.22% (w/w) in formulation induced the formation of nanosuspensions with the coexistence of amorphous and crystalline drug. This new nanosuspension formulation was related to the fact that the protein-drug complex exhibited a much stronger affinity for the drug particles over the protein itself. Interestingly, via the transcytosis pathway, the STP-Ns penetrated across the intestinal barrier into the systemic circulation, with the duodenum as the predominant absorption site. The bioavailability of the STP-Ns was 4-fold as great as that of raw crystals. The discovery of this mechanism for the use of globular protein as a stabilizer for nanosuspensions provides a new route for the preparation of amorphous drug nanoparticles. This work offers a new strategy to widen the application of globular protein and nanosuspensions of insoluble active compounds in drug delivery.
Collapse
Affiliation(s)
- Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ren L, Liu X, Wang Q, Zhang L, Deng G, Zhou F, Lu J. Facile fabrication of a magnetically smart PTX-loaded Cys–Fe3O4/CuS@BSA nano-drug for imaging-guided chemo-photothermal therapy. Dalton Trans 2017; 46:2204-2213. [DOI: 10.1039/c6dt04308a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A PTX-loaded Cys-Fe3O4/CuS@BSA nano-drug was synthesized for MR and NIR imaging-guided chemo-photothermal combination therapy of cancer via a facile fabrication method.
Collapse
Affiliation(s)
- Lanfang Ren
- School of Chemistry and Chemical Engineering
- Shanghai University of Engineering Science
- Shanghai 201620
- P. R. China
| | - Xijian Liu
- School of Chemistry and Chemical Engineering
- Shanghai University of Engineering Science
- Shanghai 201620
- P. R. China
| | - Qian Wang
- Orthopedic Traumatology
- Trauma Center
- Shanghai First People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200080
| | - Lijuan Zhang
- School of Chemistry and Chemical Engineering
- Shanghai University of Engineering Science
- Shanghai 201620
- P. R. China
| | - Guoying Deng
- Orthopedic Traumatology
- Trauma Center
- Shanghai First People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200080
| | - Feng Zhou
- Orthopedic Traumatology
- Trauma Center
- Shanghai First People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200080
| | - Jie Lu
- School of Chemistry and Chemical Engineering
- Shanghai University of Engineering Science
- Shanghai 201620
- P. R. China
| |
Collapse
|
36
|
Zhou Z, Yan J, Sun T, Wang X, Xie Z. Nanoprodrug of retinoic acid-modified paclitaxel. Org Biomol Chem 2017; 15:9611-9615. [PMID: 29106434 DOI: 10.1039/c7ob02553j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A nanoprodrug with high content (75%) and increased water solubility of paclitaxel was prepared from retinoic acid-modified paclitaxel.
Collapse
Affiliation(s)
- Zijun Zhou
- Jilin Cancer Hospital
- Changchun
- P. R. China
| | | | - Tingting Sun
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- P. R. China
| | - Xin Wang
- Department of Thyroid Surgery
- The First Hospital of Jilin University
- Changchun
- P. R. China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- P. R. China
| |
Collapse
|
37
|
Ferrari R, Talamini L, Violatto MB, Giangregorio P, Sponchioni M, Morbidelli M, Salmona M, Bigini P, Moscatelli D. Biocompatible Polymer Nanoformulation To Improve the Release and Safety of a Drug Mimic Molecule Detectable via ICP-MS. Mol Pharm 2016; 14:124-134. [DOI: 10.1021/acs.molpharmaceut.6b00753] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Raffaele Ferrari
- Institute
for Chemical and Bioengineering, Department of Chemistry and Applied
Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1, 8093 Zurich, Switzerland
| | - Laura Talamini
- Department
of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156 Milan, Italy
| | - Martina Bruna Violatto
- Department
of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156 Milan, Italy
| | - Paola Giangregorio
- Department
of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156 Milan, Italy
| | - Mattia Sponchioni
- Department
of Chemistry, Materials and Chemical Engineering, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy
| | - Massimo Morbidelli
- Institute
for Chemical and Bioengineering, Department of Chemistry and Applied
Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1, 8093 Zurich, Switzerland
| | - Mario Salmona
- Department
of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156 Milan, Italy
| | - Paolo Bigini
- Department
of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156 Milan, Italy
| | - Davide Moscatelli
- Department
of Chemistry, Materials and Chemical Engineering, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy
| |
Collapse
|
38
|
Arranja AG, Pathak V, Lammers T, Shi Y. Tumor-targeted nanomedicines for cancer theranostics. Pharmacol Res 2016; 115:87-95. [PMID: 27865762 DOI: 10.1016/j.phrs.2016.11.014] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/25/2016] [Accepted: 11/15/2016] [Indexed: 11/26/2022]
Abstract
Chemotherapeutic drugs have multiple drawbacks, including severe side effects and suboptimal therapeutic efficacy. Nanomedicines assist in improving the biodistribution and target accumulation of chemotherapeutic drugs, and are therefore able to enhance the balance between efficacy and toxicity. Multiple types of nanomedicines have been evaluated over the years, including liposomes, polymer-drug conjugates and polymeric micelles, which rely on strategies such as passive targeting, active targeting and triggered release for improved tumor-directed drug delivery. Based on the notion that tumors and metastases are highly heterogeneous, it is important to integrate imaging properties in nanomedicine formulations in order to enable non-invasive and quantitative assessment of targeting efficiency. By allowing for patient pre-selection, such next generation nanotheranostics are useful for facilitating clinical translation and personalizing nanomedicine treatments.
Collapse
Affiliation(s)
- Alexandra G Arranja
- Department of Chemical Engineering, Delft University of Technology, 2628BL Delft, The Netherlands
| | - Vertika Pathak
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, 52074 Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, 52074 Aachen, Germany.,Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands.,Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Yang Shi
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, 52074 Aachen, Germany
| |
Collapse
|