101
|
Patnaik S. Nanomedicine Magic Bullet for Human Cancer. Oncology 2017. [DOI: 10.4018/978-1-5225-0549-5.ch014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nanotechnology is the new tool that has changed healthcare, engineering, and space science. The technology involves nanoparticles that are effectively a bridge between bulk materials and atomic or molecular structures. The properties of materials change its surface plasmon resonance in metals, supermagnetism in magnetic materials as their size approaches to nanoscale. Taking in to account of their small sizes (less than 100nm) and their miraculous properties, unlike their precursor bulk material, nanoparticles are exploited to create new diagnostics and therapeutics with respect to several human diseases. Nanomedicine is generating a new generation of innovative revolution in nanoscale drug delivery strategies, site-specific drug delivery, and personalized therapy in cancer by releasing the drug at a specific site. This chapter discusses the evolution of nanomedicine to several advancements in the field of nanoparticle technologies, targeting and controlled release strategies, with the desire of generating robust and efficient nanotherapeutic tools against cancer.
Collapse
|
102
|
Bayir E, Bilgi E, Urkmez AS. Implementation of Nanoparticles in Cancer Therapy. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Cancer is a wide group of diseases and generally characterized by uncontrolled proliferation of cells whose metabolic activities are disrupted. Conventionally, chemotherapy, radiotherapy, and surgery are used in the treatment of cancer. However, in theory, even a single cancer cell may trigger recurrence. Therefore, these treatments cannot provide high survival rate for deadly types. Identification of alternative methods in treatment of cancers is inevitable because of adverse effects of conventional methods. In the last few decades, nanotechnology developed by scientists working in different disciplines—physics, chemistry, and biology—offers great opportunities. It is providing elimination of both circulating tumor cells and solid cancer cells by targeting cancer cells. In this chapter, inadequate parts of conventional treatment methods, nanoparticle types used in new treatment methods of cancer, and targeting methods of nanoparticles are summarized; furthermore, recommendations of future are provided.
Collapse
|
103
|
Lee SW, Kim YM, Kim YT, Kang SB. An open-label, multicenter, phase I trial of a cremophor-free, polymeric micelle formulation of paclitaxel combined with carboplatin as a first-line treatment for advanced ovarian cancer: a Korean Gynecologic Oncology Group study (KGOG-3016). J Gynecol Oncol 2016; 28:e26. [PMID: 28028994 PMCID: PMC5391390 DOI: 10.3802/jgo.2017.28.e26] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/17/2016] [Accepted: 12/08/2016] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE This phase I study aimed to determine the maximum tolerated dose (MTD) of Genexol-PM, when combined with carboplatin, as a first-line treatment in patients with advanced ovarian cancer. METHODS This open-label, multicenter, phase I, dose-escalation study included 18 patients (median age: 59.0 years, range: 40-75 years) diagnosed with advanced epithelial ovarian cancer. All patients had measurable residual disease after debulking surgery. Patients were assigned to groups (n=6 each group) that received different doses of Genexol-PM (220, 260, and 300 mg/m², once every 3 weeks) and 5 area under the curve (AUC) carboplatin. Safety and efficacy were analyzed for each dose group. RESULTS In this intention-to-treat population, 3 out of 18 patients dropped out of the study: 1 due to dose-limiting toxicity (DLT), 1 due to hypersensitivity, and 1 was lost during follow-up. DLTs were not reported at 220 mg/m² or 260 mg/m², but at 300 mg/m², 1 patient experienced DLT (grade 3 general pain). The MTD of Genexol-PM was not determined, but a dose of 300 mg/m² or less could be recommended for the phase II study. Most patients (73.9%) with adverse events recovered without sequelae, and no death occurred that was related to the disease or treatment. The best overall response rate was 94.1%. CONCLUSION Genexol-PM combined with carboplatin was well tolerated as a first-line treatment, and good responses were observed in patients with advanced ovarian cancer. Based on these results, we recommended a dose of 300 mg/m² or less for a phase II study.
Collapse
Affiliation(s)
- Shin Wha Lee
- Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yong Man Kim
- Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| | - Young Tae Kim
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University, Seoul, Korea
| | - Soon Beom Kang
- Department of Obstetrics and Gynecology, Konkuk University Medical Center, Seoul, Korea
| |
Collapse
|
104
|
Jain A, Kunduru KR, Basu A, Mizrahi B, Domb AJ, Khan W. Injectable formulations of poly(lactic acid) and its copolymers in clinical use. Adv Drug Deliv Rev 2016; 107:213-227. [PMID: 27423636 DOI: 10.1016/j.addr.2016.07.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/04/2016] [Accepted: 07/05/2016] [Indexed: 10/21/2022]
Abstract
Poly(lactic acid) and its copolymers have revolutionized the field of drug delivery due to their excellent biocompatibility and tunable physico-chemical properties. These copolymers have served the healthcare sector by contributing many products to combat various diseases and for biomedical applications. This article provides a comprehensive overview of clinically used products of poly(lactic acid) and its copolymers. Multi-dimension information covering product approval, formulation aspects and clinical status is described to provide a panoramic overview of each product. Moreover, leading patented technologies and various clinical trials on these products for different applications are included. This review focuses on marketed injectable formulations of PLA and its copolymers.
Collapse
|
105
|
Zhang L, Tan L, Chen L, Chen X, Long C, Peng J, Qian Z. A simple method to improve the stability of docetaxel micelles. Sci Rep 2016; 6:36957. [PMID: 27833135 PMCID: PMC5105067 DOI: 10.1038/srep36957] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/24/2016] [Indexed: 02/05/2023] Open
Abstract
Self-assembled polymeric micelles have been widely applied in drug delivery systems. In this study, we found that pH value of micellar system solution was the decisive factor of physical stability. Furthermore, the weak basic solution could maintain the solution clarification for a relative long time. To investigate the stability of polymeric micelles in different pH solutions, the micellar particle size and the docetaxel content remaining in solution were detected at predetermined time points. The crystallographic assay of freeze-drying powder was characterized by an X-ray diffractometer. In vitro release results indicated that the PBS had little influence on the sustained-release effect of docetaxel-loaded polymeric micelles (DPM). Besides, the safety of micellar formulation was determined by an MTT assay on HEK293 cells, and the anti-tumor activity was tested on MCF-7 cells. The results demonstrated that DPM adjusted with PBS (DPM (PBS)) was of low toxicity and maintained the effectiveness of docetaxel. In vivo antitumor results indicated that DPM (PBS) had better antitumor efficacy than common docetaxel injection (DTX). Thus it was concluded that regulation of micellar solution PH by PBS is a safe and effective method to improve the physical stability of DPM. It might promote the application of micellar formulation in clinical applications.
Collapse
Affiliation(s)
- Lan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China.,Research and Development Department, Guangdong Zhongsheng Pharmacy, Dongguan, 523325, China
| | - LiWei Tan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - LiJuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - XiaoXin Chen
- Research and Development Department, Guangdong Zhongsheng Pharmacy, Dongguan, 523325, China
| | - ChaoFeng Long
- Research and Development Department, Guangdong Zhongsheng Pharmacy, Dongguan, 523325, China
| | - JinRong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - ZhiYong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
106
|
Chowdhury S, Yusof F, Salim WWAW, Sulaiman N, Faruck MO. An overview of drug delivery vehicles for cancer treatment: Nanocarriers and nanoparticles including photovoltaic nanoparticles. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 164:151-159. [DOI: 10.1016/j.jphotobiol.2016.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/09/2016] [Accepted: 09/10/2016] [Indexed: 02/02/2023]
|
107
|
Synthesis, characterization and drug loading property of Monomethoxy-Poly(ethylene glycol)-Poly(ε-caprolactone)-Poly(D,L-lactide) (MPEG-PCLA) copolymers. Sci Rep 2016; 6:34069. [PMID: 27677842 PMCID: PMC5039728 DOI: 10.1038/srep34069] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/07/2016] [Indexed: 02/05/2023] Open
Abstract
Amphiphilic block copolymers have attracted a great deal of attention in drug delivery systems. In this work, a series of monomethoxy-poly (ethylene glycol)-poly (ε-caprolactone-co-D,L-lactide) (MPEG-PCLA) copolymers with variable composition of poly (ε-caprolactone) (PCL) and poly (D,L-lactide) (PDLLA) were prepared via ring-opening copolymerization of ε-CL and D,L-LA in the presence of MPEG and stannous octoate. The structure and molecular weight were characterized by nuclear magnetic resonance (NMR) and gel permeation chromatography (GPC). The crystallinity, hydrophilicity, thermal stability and hydrolytic degradation behavior were investigated in detail, respectively. The results showed that the prepared amphiphilic MPEG-PCLA copolymers have adjustable properties by altering the composition of PCLA, which make it convenient for clinical applications. Besides, the drug loading properties were also studied. Docetaxel (DTX) could be entrapped in MPEG-PCLA micelles with high loading capacity and encapsulation efficiency. And all lyophilized DTX-loaded MPEG-PCLA micelles except MPEG-PCL micelles were readily re-dissolved in normal saline at 25 °C. In addition, DTX-loaded MPEG-PCLA micelles showed a slightly enhanced antitumor activity compared with free DTX. Furthermore, DTX micelles exhibited a slower and sustained release behavior in vitro, and higher DTX concentration and longer retention time in vivo. The results suggested that the MPEG-PCLA copolymer with the adjustable ratio of PCL to PDLLA may be a promising drug delivery carrier for DTX.
Collapse
|
108
|
|
109
|
Nishiyama N, Matsumura Y, Kataoka K. Development of polymeric micelles for targeting intractable cancers. Cancer Sci 2016; 107:867-74. [PMID: 27116635 PMCID: PMC4946707 DOI: 10.1111/cas.12960] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/16/2022] Open
Abstract
In relation to recent advances in nanobiotechnologies, cancer-targeted therapy using nano-scaled drug carriers (nanocarriers) has been attracting enormous attention with success in clinical studies. Polymeric micelles, core-shell-type nanoparticles formed through the self-assembly of block copolymers, are one of the most promising nanocarrier, because their critical features such as size, stability, and drug incorporation efficiency and release rate can be modulated by designing the constituent block copolymers. The utilities of polymeric micelles have been reported not only in experimental tumor models in mice but also in clinical studies. In this article, we aim to explain the rationale of designing polymeric micelles for targeting intractable cancers such as pancreatic cancer, glioblastoma, and metastases. Also, we review recent progress in clinical studies on polymeric micelles incorporating anticancer drugs. In addition, we introduce the next generation of polymeric micelles as the platform integrated with smart functionalities such as targetability, environmental sensitivity, and imaging properties. Thus, polymeric micelles can realize safe and effective cancer therapy, and offer tailor-made medicines for individual patients.
Collapse
Affiliation(s)
- Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.,Innovation Center of NanoMedicine (iCONM), Kawasaki, Japan
| | - Yasuhiro Matsumura
- Innovation Center of NanoMedicine (iCONM), Kawasaki, Japan.,Investigative Treatment Division, Research Center for Innovative Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki, Japan.,Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
110
|
Caster JM, Patel AN, Zhang T, Wang A. Investigational nanomedicines in 2016: a review of nanotherapeutics currently undergoing clinical trials. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27312983 DOI: 10.1002/wnan.1416] [Citation(s) in RCA: 229] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/09/2016] [Accepted: 05/17/2016] [Indexed: 12/16/2022]
Abstract
Nanomedicine is a relatively new field that is rapidly evolving. Formulation of drugs on the nanoscale imparts many physical and biological advantages. Such advantages can in turn translate into improved therapeutic efficacy and reduced toxicity. While approximately 50 nanotherapeutics have already entered clinical practice, a greater number of drugs are undergoing clinical investigation for a variety of indications. This review aims to examine all the nanoformulations that are currently undergoing clinical investigation and their outlook for ultimate clinical translation. WIREs Nanomed Nanobiotechnol 2017, 9:e1416. doi: 10.1002/wnan.1416 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Joseph M Caster
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Artish N Patel
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tian Zhang
- Department of Medicine, Duke University, Durham, NC, USA
| | - Andrew Wang
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
111
|
Zhang Y, Buhrman JS, Liu Y, Rayahin JE, Gemeinhart RA. Reducible Micelleplexes are Stable Systems for Anti-miRNA Delivery in Cerebrospinal Fluid. Mol Pharm 2016; 13:1791-9. [PMID: 27177352 DOI: 10.1021/acs.molpharmaceut.5b00933] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glioblastoma multiforme (GBM) and other central nervous system (CNS) cancers have poor long-term prognosis, and there is a significant need for improved treatments. GBM initiation and progression are mediated, in part, by microRNA (miRNA), which are endogenous posttranscriptional gene regulators. Misregulation of miRNAs is a potential target for therapeutic intervention in GBM. In this work, a micelle-like nanoparticle delivery system based upon the block copolymer poly(ethylene glycol-b-lactide-b-arginine) was designed with and without a reducible linkage between the lactide and RNA-binding peptide, R15, to assess the ability of the micelle-like particles to disassemble. Using confocal live cell imaging, intracellular dissociation was pronounced for the reducible micelleplexes. This dissociation was also supported by higher efficiency in a dual luciferase assay specific for the miRNA of interest, miR-21. Notably, micelleplexes were found to have significantly better stability and higher anti-miRNA activity in cerebrospinal fluid than in human plasma, suggesting an advantage for applying micelleplexes to CNS diseases and in vivo CNS therapeutics. The reducible delivery system was determined to be a promising delivery platform for the treatment of CNS diseases with miRNA therapy.
Collapse
Affiliation(s)
| | | | | | | | - Richard A Gemeinhart
- Department of Bioengineering, University of Illinois , Chicago, Illinois 60607-7052, United States
| |
Collapse
|
112
|
Digesu CS, Hofferberth SC, Grinstaff MW, Colson YL. From Diagnosis to Treatment: Clinical Applications of Nanotechnology in Thoracic Surgery. Thorac Surg Clin 2016; 26:215-28. [PMID: 27112260 PMCID: PMC4851727 DOI: 10.1016/j.thorsurg.2015.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nanotechnology is an emerging field with potential as an adjunct to cancer therapy, particularly thoracic surgery. Therapy can be delivered to tumors in a more targeted fashion, with less systemic toxicity. Nanoparticles may aid in diagnosis, preoperative characterization, and intraoperative localization of thoracic tumors and their lymphatics. Focused research into nanotechnology's ability to deliver both diagnostics and therapeutics has led to the development of nanotheranostics, which promises to improve the treatment of thoracic malignancies through enhanced tumor targeting, controlled drug delivery, and therapeutic monitoring. This article reviews nanoplatforms, their unique properties, and the potential for clinical application in thoracic surgery.
Collapse
Affiliation(s)
- Christopher S Digesu
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, 15 Francis St, Boston, MA 02115, USA
| | - Sophie C Hofferberth
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, 15 Francis St, Boston, MA 02115, USA
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Metcalf Science Center, Boston University, SCI 518, 590 Commonwealth Avenue, Boston, MA 02215, USA; Department of Chemistry, Metcalf Science Center, Boston University, SCI 518, 590 Commonwealth Avenue, Boston, MA 02215, USA; Department of Medicine, Metcalf Science Center, Boston University, SCI 518, 590 Commonwealth Avenue, Boston, MA 02215, USA
| | - Yolonda L Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, 15 Francis St, Boston, MA 02115, USA; Division of Thoracic Surgery, Brigham and Women's Hospital, Harvard Medical School, 15 Francis St, Boston, MA 02155, USA.
| |
Collapse
|
113
|
Xu CF, Zhang HB, Sun CY, Liu Y, Shen S, Yang XZ, Zhu YH, Wang J. Tumor acidity-sensitive linkage-bridged block copolymer for therapeutic siRNA delivery. Biomaterials 2016; 88:48-59. [PMID: 26945455 DOI: 10.1016/j.biomaterials.2016.02.031] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/14/2016] [Accepted: 02/19/2016] [Indexed: 10/22/2022]
Abstract
The design of ideal nanoparticle delivery systems should be capable of meeting the requirements of several stages of drug delivery, including prolonged circulation, enhanced accumulation and penetration in the tumor, facilitated cellular internalization and rapid release of the active drug in the tumor cells. However, among the current design strategies, meeting the requirements of one stage often conflicts with the other. Herein, a tumor pH-labile linkage-bridged block copolymer of poly(ethylene glycol) with poly(lacide-co-glycolide) (PEG-Dlinkm-PLGA) was used for siRNA delivery to fulfill all aforementioned requirements of these delivery stages. The obtained siRNA-encapsulating PEG-Dlinkm-PLGA nanoparticle gained efficiently prolonged circulation in the blood and preferential accumulation in tumor sites via the PEGylation. Furthermore, the PEG surface layer was detached in response to the tumor acidic microenvironment to facilitate cellular uptake, and the siRNA was rapidly released within tumor cells due to the hydrophobic PLGA layer. Hence, PEG-Dlinkm-PLGA nanoparticles met the requirements of several stages of drug delivery, and resulted in the enhanced therapeutic effect of the nanoparticular delivery systems.
Collapse
Affiliation(s)
- Cong-Fei Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei Anhui 230027, PR China
| | - Hou-Bing Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei Anhui 230027, PR China
| | - Chun-Yang Sun
- CAS Center for Excellence in Nanoscience, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei Anhui 230027, PR China
| | - Yang Liu
- CAS Center for Excellence in Nanoscience, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei Anhui 230027, PR China
| | - Song Shen
- CAS Center for Excellence in Nanoscience, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei Anhui 230027, PR China
| | - Xian-Zhu Yang
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei Anhui 230027, PR China.
| | - Yan-Hua Zhu
- CAS Center for Excellence in Nanoscience, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei Anhui 230027, PR China
| | - Jun Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei Anhui 230027, PR China; CAS Center for Excellence in Nanoscience, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei Anhui 230027, PR China; Innovation Center for Cell Signaling Network, University of Science and Technology of China, Hefei, Anhui 230027, PR China.
| |
Collapse
|
114
|
Ulbrich K, Holá K, Šubr V, Bakandritsos A, Tuček J, Zbořil R. Targeted Drug Delivery with Polymers and Magnetic Nanoparticles: Covalent and Noncovalent Approaches, Release Control, and Clinical Studies. Chem Rev 2016; 116:5338-431. [DOI: 10.1021/acs.chemrev.5b00589] [Citation(s) in RCA: 1086] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Karel Ulbrich
- Institute
of Macromolecular Chemistry, The Czech Academy of Sciences, v.v.i., Heyrovsky Square 2, 162 06 Prague 6, Czech Republic
| | - Kateřina Holá
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Vladimir Šubr
- Institute
of Macromolecular Chemistry, The Czech Academy of Sciences, v.v.i., Heyrovsky Square 2, 162 06 Prague 6, Czech Republic
| | - Aristides Bakandritsos
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Jiří Tuček
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Radek Zbořil
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| |
Collapse
|
115
|
Yang J, Li Y, Zhang T, Zhang X. Development of bioactive materials for glioblastoma therapy. Bioact Mater 2016; 1:29-38. [PMID: 29744393 PMCID: PMC5883963 DOI: 10.1016/j.bioactmat.2016.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma is the most common and deadly human brain cancers. Unique barriers hinder the drug delivering pathway due to the individual position of glioblastoma, including blood-brain barrier and blood-brain tumor barrier. Numerous bioactive materials have been exploited and applied as the transvascular delivery carriers of therapeutic drugs. They promote site-specific accumulation and long term release of the encapsulated drugs at the tumor sites and reduce side effects with systemic delivery. And the delivery systems exhibit a certain extent of anti-glioblastoma effect and extend the median survival time. However, few of them step into the clinical trials. In this review, we will investigate the recent studies of bioactive materials for glioblastoma chemotherapy, including the inorganic materials, lipids and polymers. These bioactive materials construct diverse delivery vehicles to trigger tumor sites in brain intravenously. Herein, we exploit their functionality in drug delivery and discuss the deficiency for the featured tumors, to provide guidance for establishing optimized therapeutic drug formulation for anti-glioblastoma therapy and pave the way for clinical application. Numerous bioactive materials have been exploited as delivery carriers of therapeutic drugs for glioblastoma chemotherapy. The functionality and deficiency of the bioactive materials are discussed. Combing the chemo- and immunotherapy will provide a promising strategy for glioblastoma therapy and inhibiting recurrence.
Collapse
Key Words
- ALA, α-lipoic acid
- BAG3, Bcl-2 associated athanogene 3
- BBB, blood-brain barrier
- BTB, blood-brain tumor barrier
- Bioactive material
- Blood-brain barrier
- Blood-brain tumor barrier
- CNS, central nervous system
- CPT, camptothecin
- Chemotherapy
- DACHPt, dichloro-(1,2-diaminocyclohexane)platinum (II)
- DCs, dendritic cells
- DHA, dehydroascorbic acid
- DOX, doxorubicin
- DPPC, 1,2-dihexadecanoyl-rac-glycero-3-phosphocholine
- FA, folate
- GCV, ganciclovir
- GLUT1, glucose transporter isoform 1
- Glioblastoma
- IL, interleukin
- MMPs, matrix metalloproteinases
- PTX, paclitaxel
- ROS, reactive oxygen species
- SN38, 7-ethyl-10-hydroxy-camptothecin
- TAT, transactivator of transcription
- TEG, tetra(ethylene glycol)
- TMZ, temozolomide
- TNF, tumor necrosis factor
- TfR, transferrin receptor
- cRGD, cyclic Arg-Gly-Asp
Collapse
Affiliation(s)
- Jun Yang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yan Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Tianlu Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xin Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
116
|
Xie J, Yang Z, Zhou C, Zhu J, Lee RJ, Teng L. Nanotechnology for the delivery of phytochemicals in cancer therapy. Biotechnol Adv 2016; 34:343-353. [PMID: 27071534 DOI: 10.1016/j.biotechadv.2016.04.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 03/01/2016] [Accepted: 04/07/2016] [Indexed: 02/06/2023]
Abstract
The aim of this review is to summarize advances that have been made in the delivery of phytochemicals for cancer therapy by the use of nanotechnology. Over recent decades, much research effort has been invested in developing phytochemicals as cancer therapeutic agents. However, several impediments to their wide spread use as drugs still have to be overcome. Among these are low solubility, poor penetration into cells, high hepatic disposition, and narrow therapeutic index. Rapid clearance or uptake by normal tissues and wide tissue distribution result in low drug accumulation in the target tumor sites can result in undesired drug exposure in normal tissues. Association with or encapsulation in nanoscale drug carriers is a potential strategy to address these problems. This review discussed lessons learned on the use of nanotechnology for delivery of phytochemicals that been tested in clinical trials or are moving towards the clinic.
Collapse
Affiliation(s)
- Jing Xie
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Zhaogang Yang
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus OH 43210, USA
| | - Chenguang Zhou
- Department of Pharmacokinetics and Pharmacodynamics, Genentech, San Francisco 94080, CA, USA
| | - Jing Zhu
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus OH 43210, USA
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun 130012, China; Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus OH 43210, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
117
|
Wang K, Yuan A, Yu J, Wu J, Hu Y. One-Step Self-Assembling Method to Prepare Dual-Functional Transferrin Nanoparticles for Antitumor Drug Delivery. J Pharm Sci 2016; 105:1269-76. [DOI: 10.1016/j.xphs.2015.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 12/01/2015] [Accepted: 12/08/2015] [Indexed: 01/13/2023]
|
118
|
Abstract
PURPOSE OF REVIEW Recent advances in nanotechnology have addressed some of the issues related to lack of selectivity and nonspecific toxicities associated with conventional chemotherapy. Nanoparticles are therapeutic carriers that can be fine tuned for specific application and for passive or active tumor targeting. RECENT FINDINGS Although the nanoparticle field is rapidly expanding, there are to date only six nanoparticle-based drug delivery platforms and two antibody-drug conjugates that are clinically approved for cancer therapy. Here, we review the clinical data of liposomal anthracyclines, nanoparticle formulations of paclitaxel and trastuzumab emtansine. We then briefly comment on efficacy and safety issues of nanoparticles, as well as on the next-generation nanoparticles for cancer therapy. SUMMARY The emerging development of cancer nanotechnology offers the opportunity of reinvestigating the potential of cytotoxic agents, improving tumor targeting and drug delivery, leading to better safety profile and antitumor activity. Adding specificity to nanoparticles may allow personalization of cancer therapy using chemotherapy.
Collapse
|
119
|
Rodzinski A, Guduru R, Liang P, Hadjikhani A, Stewart T, Stimphil E, Runowicz C, Cote R, Altman N, Datar R, Khizroev S. Targeted and controlled anticancer drug delivery and release with magnetoelectric nanoparticles. Sci Rep 2016; 6:20867. [PMID: 26875783 PMCID: PMC4753509 DOI: 10.1038/srep20867] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/08/2016] [Indexed: 12/21/2022] Open
Abstract
It is a challenge to eradicate tumor cells while sparing normal cells. We used magnetoelectric nanoparticles (MENs) to control drug delivery and release. The physics is due to electric-field interactions (i) between MENs and a drug and (ii) between drug-loaded MENs and cells. MENs distinguish cancer cells from normal cells through the membrane's electric properties; cancer cells have a significantly smaller threshold field to induce electroporation. In vitro and in vivo studies (nude mice with SKOV-3 xenografts) showed that (i) drug (paclitaxel (PTX)) could be attached to MENs (30-nm CoFe2O4@BaTiO3 nanostructures) through surface functionalization to avoid its premature release, (ii) drug-loaded MENs could be delivered into cancer cells via application of a d.c. field (~100 Oe), and (iii) the drug could be released off MENs on demand via application of an a.c. field (~50 Oe, 100 Hz). The cell lysate content was measured with scanning probe microscopy and spectrophotometry. MENs and control ferromagnetic and polymer nanoparticles conjugated with HER2-neu antibodies, all loaded with PTX were weekly administrated intravenously. Only the mice treated with PTX-loaded MENs (15/200 μg) in a field for three months were completely cured, as confirmed through infrared imaging and post-euthanasia histology studies via energy-dispersive spectroscopy and immunohistochemistry.
Collapse
Affiliation(s)
- Alexandra Rodzinski
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, USAFlorida 33199, USA
| | - Rakesh Guduru
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, USAFlorida 33199, USA
| | - Ping Liang
- Electrical and Computer Engineering, University of California, Riverside, CA 92521, USA
- Cellular Nanomed Inc., Weston, FL 33331, USA
| | - Ali Hadjikhani
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| | - Tiffanie Stewart
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, USAFlorida 33199, USA
| | - Emmanuel Stimphil
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| | - Carolyn Runowicz
- Department of Obstetrics and Gynecology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - Richard Cote
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John T. Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33136, USA.
| | - Norman Altman
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ram Datar
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John T. Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33136, USA.
| | - Sakhrat Khizroev
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, USAFlorida 33199, USA
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| |
Collapse
|
120
|
Palao-Suay R, Gómez-Mascaraque L, Aguilar M, Vázquez-Lasa B, Román JS. Self-assembling polymer systems for advanced treatment of cancer and inflammation. Prog Polym Sci 2016. [DOI: 10.1016/j.progpolymsci.2015.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
121
|
Singh RP, Sharma G, Sonali, Agrawal P, Pandey BL, Koch B, Muthu MS. Transferrin receptor targeted PLA-TPGS micelles improved efficacy and safety in docetaxel delivery. Int J Biol Macromol 2016; 83:335-44. [DOI: 10.1016/j.ijbiomac.2015.11.081] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 12/19/2022]
|
122
|
Huang WC, Burnouf PA, Su YC, Chen BM, Chuang KH, Lee CW, Wei PK, Cheng TL, Roffler SR. Engineering Chimeric Receptors To Investigate the Size- and Rigidity-Dependent Interaction of PEGylated Nanoparticles with Cells. ACS NANO 2016; 10:648-662. [PMID: 26741147 DOI: 10.1021/acsnano.5b05661] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Attachment of ligands to the surface of nanoparticles (NPs) is an attractive approach to target specific cells and increase intracellular delivery of nanocargos. To expedite investigation of targeted NPs, we engineered human cancer cells to express chimeric receptors that bind polyethylene glycol (PEG) and internalize stealth NPs in a fashion similar to ligand-targeted liposomes against epidermal growth factor receptor 1 or 2 (HER1 or HER2), which are validated targets for cancer therapy. Measurement of the rate of endocytosis and lysosomal accumulation of small (80-94 nm) or large (180-220 nm) flexible liposomes or more rigid lipid-coated mesoporous silica particles in human HT29 colon cancer and SKBR3 breast cancer cells that express chimeric receptors revealed that larger and more rigid NPs were internalized more slowly than smaller and more flexible NPs. An exception is when both the small and large liposomes underwent endocytosis via HER2. HER1 mediated faster and greater uptake of NPs into cells but retained NPs less well as compared to HER2. Lysosomal accumulation of NPs internalized via HER1 was unaffected by NP rigidity but was inversely related to NP size, whereas large rigid NPs internalized by HER2 displayed increased lysosomal accumulation. Our results provide insight into the effects of NP properties on receptor-mediated endocytosis and suggest that anti-PEG chimeric receptors may help accelerate investigation of targeted stealth NPs.
Collapse
Affiliation(s)
- Wei-Chiao Huang
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
| | - Pierre-Alain Burnouf
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica , Taipei, Taiwan
| | - Yu-Cheng Su
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University , Taipei 110, Taiwan
| | - Chia-Wei Lee
- Research Center for Applied Sciences, Academia Sinica , Taipei 11529, Taiwan
| | - Pei-Kuen Wei
- Research Center for Applied Sciences, Academia Sinica , Taipei 11529, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical and Environmental Biology, Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
| |
Collapse
|
123
|
Structural modifications in polymeric micelles to impart multifunctionality for improved drug delivery. Ther Deliv 2016; 7:73-87. [PMID: 26769002 DOI: 10.4155/tde.15.90] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Polymeric micelles are macromolecular nanoconstructs which are formed by self-assembly of synthetic amphiphilic block copolymers. These copolymers could be chemically modified to expand their functionality and hence obtain a multifunctional micelle which could serve several functions simultaneously, for example, long circulation time along with active targeting, smart polymeric micelles providing on-demand drug release for example, pH responsive micelles, redox- and light-sensitive micelles, charge-conversion micelles and core/shell cross-linked micelles. Additionally, micelles could be tailored to carry a contrast agent or siRNA/miRNA along with the drug for greater clinical benefit. The focus of the current commentary would be to highlight such chemical modifications which impart multifunctionality to a single carrier and discuss challenges involved in clinical translation of these multifunctional micelles.
Collapse
|
124
|
Du N, Guo W, Yu Q, Guan S, Guo L, Shen T, Tang H, Gan Z. Poly(d,l-lactic acid)-block-poly(N-(2-hydroxypropyl)methacrylamide) nanoparticles for overcoming accelerated blood clearance and achieving efficient anti-tumor therapy. Polym Chem 2016. [DOI: 10.1039/c6py01113f] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The substitution of PEG with PHPMA maintained the long circulation of PDLLA-b-PEG and alleviated the accelerated blood clearance (ABC).
Collapse
Affiliation(s)
- Nan Du
- The State Key Laboratory of Organic–Inorganic Composites
- Beijing Laboratory of Biomedical Materials
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Wenxuan Guo
- The State Key Laboratory of Organic–Inorganic Composites
- Beijing Laboratory of Biomedical Materials
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Qingsong Yu
- The State Key Laboratory of Organic–Inorganic Composites
- Beijing Laboratory of Biomedical Materials
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Shuli Guan
- The State Key Laboratory of Organic–Inorganic Composites
- Beijing Laboratory of Biomedical Materials
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Linyi Guo
- The State Key Laboratory of Organic–Inorganic Composites
- Beijing Laboratory of Biomedical Materials
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Tong Shen
- The State Key Laboratory of Organic–Inorganic Composites
- Beijing Laboratory of Biomedical Materials
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Hao Tang
- The State Key Laboratory of Organic–Inorganic Composites
- Beijing Laboratory of Biomedical Materials
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Zhihua Gan
- The State Key Laboratory of Organic–Inorganic Composites
- Beijing Laboratory of Biomedical Materials
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| |
Collapse
|
125
|
Luk BT, Zhang L. Cell membrane-camouflaged nanoparticles for drug delivery. J Control Release 2015; 220:600-7. [PMID: 26210440 PMCID: PMC4688192 DOI: 10.1016/j.jconrel.2015.07.019] [Citation(s) in RCA: 410] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/16/2015] [Accepted: 07/17/2015] [Indexed: 12/15/2022]
Abstract
Nanoparticles can preferentially accumulate at sites of action and hold great promise to improve the therapeutic index of many drugs. While conventional methods of nanocarrier-mediated drug delivery have focused on primarily synthetic approaches, engineering strategies that combine synthetic nanoparticles with natural biomaterials have recently gained much attention. In particular, cell membrane-camouflaged nanoparticles are a new class of biomimetic nanoparticles that combine the unique functionalities of cellular membranes and engineering versatility of synthetic nanomaterials for effective delivery of therapeutic agents. Herein, we report on the recent progress on cell membrane-coated nanoparticles for drug delivery. In particular, we highlight three areas: (i) prolonging systemic circulation via cell membrane coating, (ii) cell-specific targeting via cell membrane coating, and (iii) applications of cell membrane coating for drug delivery. The cell membrane-camouflaged nanoparticle platform has emerged as a novel delivery strategy with the potential to improve the therapeutic efficacy for the treatment of a variety of diseases.
Collapse
Affiliation(s)
- Brian T Luk
- Department of NanoEngineering, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, United States
| | - Liangfang Zhang
- Department of NanoEngineering, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
126
|
Dalela M, Shrivastav TG, Kharbanda S, Singh H. pH-Sensitive Biocompatible Nanoparticles of Paclitaxel-Conjugated Poly(styrene-co-maleic acid) for Anticancer Drug Delivery in Solid Tumors of Syngeneic Mice. ACS APPLIED MATERIALS & INTERFACES 2015; 7:26530-26548. [PMID: 26528585 DOI: 10.1021/acsami.5b07764] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In the present study, we have synthesized poly(styrene-co-maleic anhydride), a biocompatible copolymer that was further conjugated with paclitaxel (PTX) via ester linkage and self-assembled to form poly(styrene-co-maleic acid)-paclitaxel (PSMAC-PTX) nanoparticles (NPs). The in vitro release of PTX from PSMAC-PTX NPs showed a higher release at lower pH than at the physiological pH of 7.4, confirming its pH-dependent release. The cell viability of PSMAC-PTX nanoparticles was evaluated using MTT assay. IC50 values of 9.05-18.43 ng/mL of PTX equivalent were observed in various cancer cell lines after 72 h of incubation. Confocal microscopy, Western blotting, and Flow cytometry results further supported that the cellular uptake and apoptosis of cancer cells with PSMAC-PTX NPs. Pharmacokinetic studies revealed that the conjugation of PTX to the PSMAC co-polymer not only increased the plasma and tumor C(max) of PTX but also prolonged its plasma half-life and retention in tumor via enhanced permeability and retention (EPR) effect. Administration of PSMAC-PTX NPs showed significant tumor growth inhibition with improved apoptosis effects in vivo on Ehrlich Ascites Tumor (EAT)-bearing BALB/c syngeneic mice in comparison with Taxol, without showing any cytotoxicity. On the basis of preliminary results, no subacute toxicity was observed in major organs, tissues and hematological system up to a dosage of 60 mg/kg body weight in mice. Therefore, PSMAC-PTX NPs may be considered as an alternative nanodrug delivery system for the delivery of PTX in solid tumors.
Collapse
Affiliation(s)
- Manu Dalela
- Centre for Biomedical Engineering, Indian Institute of Technology , Hauz Khas, New Delhi-110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, AIIMS , New Delhi, Ansari Nagar, New Delhi-110029, India
| | - T G Shrivastav
- Department of Reproductive Biomedicine, National Institute of Health & Family Welfare , Delhi-110067, India
| | - Surender Kharbanda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, Massachusetts 02215, United States
| | - Harpal Singh
- Centre for Biomedical Engineering, Indian Institute of Technology , Hauz Khas, New Delhi-110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, AIIMS , New Delhi, Ansari Nagar, New Delhi-110029, India
| |
Collapse
|
127
|
A Prospective Phase II Study of Cisplatin and Cremophor EL-Free Paclitaxel (Genexol-PM) in Patients with Unresectable Thymic Epithelial Tumors. J Thorac Oncol 2015; 10:1800-6. [DOI: 10.1097/jto.0000000000000692] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
128
|
Hafezi MJ, Sharif F. Brownian dynamics simulation of amphiphilic block copolymers with different tail lengths, comparison with theory and comicelles. J Mol Graph Model 2015; 62:165-173. [DOI: 10.1016/j.jmgm.2015.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/03/2015] [Accepted: 09/05/2015] [Indexed: 11/30/2022]
|
129
|
Bölükbas DA, Meiners S. Lung cancer nanomedicine: potentials and pitfalls. Nanomedicine (Lond) 2015; 10:3203-12. [PMID: 26472521 DOI: 10.2217/nnm.15.155] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is by far the most common cause of cancer-related deaths in the world. Nanoparticle-based therapies enable targeted drug delivery for lung cancer treatment with increased therapeutic efficiency and reduced systemic toxicity. At the same time, nanomedicine has the potential for multimodal treatment of lung cancer that may involve 'all-in-one' targeting of several tumor-associated cell types in a timely and spatially controlled manner. Therapeutic approaches, however, are hampered by a translational gap between basic scientists, clinicians and pharma industry due to suboptimal animal models and difficulties in scale-up production of nanoagents. This calls for a disease-centered approach with interdisciplinary basic and clinical research teams with the support of pharma industries.
Collapse
Affiliation(s)
- Deniz Ali Bölükbas
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Member of the German Center for Lung Research (DZL), Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Member of the German Center for Lung Research (DZL), Germany
| |
Collapse
|
130
|
Gothwal A, Khan I, Gupta U. Polymeric Micelles: Recent Advancements in the Delivery of Anticancer Drugs. Pharm Res 2015. [PMID: 26381278 DOI: 10.1007/s11095‐015‐1784‐1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanotechnology, in health and medicine, extensively improves the safety and efficacy of different therapeutic agents, particularly the aspects related to drug delivery and targeting. Among various nano-carriers, polymer based macromolecular approaches have resulted in improved drug delivery for the diseases like cancers, diabetes, autoimmune disorders and many more. Polymeric micelles consisting of hydrophilic exterior and hydrophobic core have established a record of anticancer drug delivery from the laboratory to commercial reality. The nanometric size, tailor made functionality, multiple choices of polymeric micelle synthesis and stability are the unique properties, which have attracted scientists and researchers around the world to work upon in this opportunistic drug carrier. The capability of polymeric micelles as nano-carriers are nowhere less significant than nanoparticles, liposomes and other nanocarriers, as per as the commercial feasibility and presence is concerned. In fact polymeric micelles are among the most extensively studied delivery platforms for the effective treatment of different cancers as well as non-cancerous disorders. The present review highlights the sequential and recent developments in the design, synthesis, characterization and evaluation of polymeric micelles to achieve the effective anticancer drug delivery. The future possibilities and clinical outcome have also been discussed, briefly.
Collapse
Affiliation(s)
- Avinash Gothwal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Iliyas Khan
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
131
|
Gothwal A, Khan I, Gupta U. Polymeric Micelles: Recent Advancements in the Delivery of Anticancer Drugs. Pharm Res 2015; 33:18-39. [PMID: 26381278 DOI: 10.1007/s11095-015-1784-1] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 08/24/2015] [Indexed: 12/24/2022]
Abstract
Nanotechnology, in health and medicine, extensively improves the safety and efficacy of different therapeutic agents, particularly the aspects related to drug delivery and targeting. Among various nano-carriers, polymer based macromolecular approaches have resulted in improved drug delivery for the diseases like cancers, diabetes, autoimmune disorders and many more. Polymeric micelles consisting of hydrophilic exterior and hydrophobic core have established a record of anticancer drug delivery from the laboratory to commercial reality. The nanometric size, tailor made functionality, multiple choices of polymeric micelle synthesis and stability are the unique properties, which have attracted scientists and researchers around the world to work upon in this opportunistic drug carrier. The capability of polymeric micelles as nano-carriers are nowhere less significant than nanoparticles, liposomes and other nanocarriers, as per as the commercial feasibility and presence is concerned. In fact polymeric micelles are among the most extensively studied delivery platforms for the effective treatment of different cancers as well as non-cancerous disorders. The present review highlights the sequential and recent developments in the design, synthesis, characterization and evaluation of polymeric micelles to achieve the effective anticancer drug delivery. The future possibilities and clinical outcome have also been discussed, briefly.
Collapse
Affiliation(s)
- Avinash Gothwal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Iliyas Khan
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
132
|
Banik BL, Fattahi P, Brown JL. Polymeric nanoparticles: the future of nanomedicine. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:271-99. [PMID: 26314803 DOI: 10.1002/wnan.1364] [Citation(s) in RCA: 255] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 07/16/2015] [Accepted: 07/22/2015] [Indexed: 12/22/2022]
Abstract
Polymeric nanoparticles (NPs) are one of the most studied organic strategies for nanomedicine. Intense interest lies in the potential of polymeric NPs to revolutionize modern medicine. To determine the ideal nanosystem for more effective and distinctly targeted delivery of therapeutic applications, particle size, morphology, material choice, and processing techniques are all research areas of interest. Utilizations of polymeric NPs include drug delivery techniques such as conjugation and entrapment of drugs, prodrugs, stimuli-responsive systems, imaging modalities, and theranostics. Cancer, neurodegenerative disorders, and cardiovascular diseases are fields impacted by NP technologies that push scientific boundaries to the leading edge of transformative advances for nanomedicine.
Collapse
Affiliation(s)
- Brittany L Banik
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Pouria Fattahi
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Justin L Brown
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
133
|
Affiliation(s)
- Yuanzeng Min
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Joseph M Caster
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Michael J Eblan
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
134
|
Graphene oxide stabilized by PLA–PEG copolymers for the controlled delivery of paclitaxel. Eur J Pharm Biopharm 2015; 93:18-26. [DOI: 10.1016/j.ejpb.2015.03.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/04/2015] [Accepted: 03/12/2015] [Indexed: 11/23/2022]
|
135
|
Chung H, Lee H, Han HK, An H, Lim KS, Lee YJ, Cho JY, Yoon SH, Jang IJ, Yu KS. A pharmacokinetic comparison of two voriconazole formulations and the effect of CYP2C19 polymorphism on their pharmacokinetic profiles. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2609-16. [PMID: 25999694 PMCID: PMC4435089 DOI: 10.2147/dddt.s80066] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Purpose SYP-1018 is a lyophilized polymeric nanoparticle formulation of voriconazole that is under development for intravenous dosing. This study compared the pharmacokinetic and tolerability profiles of SYP-1018 with those of Vfend®, the marketed formulation of voriconazole. The effect of CYP2C19 polymorphism on the voriconazole pharmacokinetics was also evaluated. Methods An open-label, two-treatment, two-period, two-sequence crossover study was conducted in 52 healthy male volunteers, who randomly received a single intravenous infusion of either of the two voriconazole formulations at 200 mg. Blood samples were collected up to 24 hours after drug administration for pharmacokinetic analysis. The plasma concentrations of voriconazole were determined using liquid chromatography with tandem mass spectrometry, and the pharmacokinetic parameters were estimated using a noncompartmental method. CYP2C19 genotype was identified in 51 subjects. Results The geometric mean ratio (90% confidence interval) of SYP-1018 to Vfend® was 0.99 (0.93–1.04) for the maximum plasma concentrations (Cmax) and 0.97 (0.92–1.01) for the area under the concentration–time curve (AUC) from dosing to the last quantifiable concentration (AUClast). Nineteen homozygous extensive metabolizers (EMs, *1/*1), 19 intermediate metabolizers (IMs, *1/*2 or *1/*3), and ten poor metabolizers (PMs, *2/*2, *2/*3, or *3/*3) were identified, and the pharmacokinetic comparability between SYP-1018 and Vfend® was also noted when analyzed separately by genotype. The systemic exposure to voriconazole was greatest in the PM group, followed by the IM, and then the EM groups. Furthermore, the intrasubject variability for Cmax and AUClast was greater in IMs and PMs than in EMs. No serious adverse event occurred, and both treatments were well tolerated. Conclusion SYP-1018 had comparable pharmacokinetic and tolerability profiles to Vfend® after a single intravenous infusion. CYP2C19 genotype affected not only the pharmacokinetics of voriconazole, but its intrasubject variability. SYP-1018 can be further developed as a clinically effective alternative to Vfend®.
Collapse
Affiliation(s)
- Hyewon Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Howard Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea ; Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hye Kyung Han
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyungmi An
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyoung Soo Lim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea ; Department of Clinical Pharmacology and Therapeutics, CHA University School of Medicine and CHA Bundang Medical Center, Seongnam, Republic of Korea
| | - Yong Jin Lee
- Medical and Regulatory Affairs Team, Samyang Biopharmaceuticals Corporation, Seoul, Republic of Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seo Hyun Yoon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
136
|
Shi Y, van der Meel R, Theek B, Blenke EO, Pieters EH, Fens MH, Ehling J, Schiffelers RM, Storm G, van Nostrum CF, Lammers T, Hennink WE. Complete Regression of Xenograft Tumors upon Targeted Delivery of Paclitaxel via Π-Π Stacking Stabilized Polymeric Micelles. ACS NANO 2015; 9:3740-52. [PMID: 25831471 PMCID: PMC4523313 DOI: 10.1021/acsnano.5b00929] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Treatment of cancer patients with taxane-based chemotherapeutics, such as paclitaxel (PTX), is complicated by their narrow therapeutic index. Polymeric micelles are attractive nanocarriers for tumor-targeted delivery of PTX, as they can be tailored to encapsulate large amounts of hydrophobic drugs and achiv prolonged circulation kinetics. As a result, PTX deposition in tumors is increased, while drug exposure to healthy tissues is reduced. However, many PTX-loaded micelle formulations suffer from low stability and fast drug release in the circulation, limiting their suitability for systemic drug targeting. To overcome these limitations, we have developed PTX-loaded micelles which are stable without chemical cross-linking and covalent drug attachment. These micelles are characterized by excellent loading capacity and strong drug retention, attributed to π-π stacking interaction between PTX and the aromatic groups of the polymer chains in the micellar core. The micelles are based on methoxy poly(ethylene glycol)-b-(N-(2-benzoyloxypropyl)methacrylamide) (mPEG-b-p(HPMAm-Bz)) block copolymers, which improved the pharmacokinetics and the biodistribution of PTX, and substantially increased PTX tumor accumulation (by more than 2000%; as compared to Taxol or control micellar formulations). Improved biodistribution and tumor accumulation were confirmed by hybrid μCT-FMT imaging using near-infrared labeled micelles and payload. The PTX-loaded micelles were well tolerated at different doses, while they induced complete tumor regression in two different xenograft models (i.e., A431 and MDA-MB-468). Our findings consequently indicate that π-π stacking-stabilized polymeric micelles are promising carriers to improve the delivery of highly hydrophobic drugs to tumors and to increase their therapeutic index.
Collapse
Affiliation(s)
- Yang Shi
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Roy van der Meel
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Benjamin Theek
- Department of Experimental Molecular Imaging (ExMI), Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Clinic, Aachen, Germany
| | - Erik Oude Blenke
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Ebel H.E. Pieters
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Marcel H.A.M. Fens
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Josef Ehling
- Department of Experimental Molecular Imaging (ExMI), Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Clinic, Aachen, Germany
| | - Raymond M. Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
- Department of Controlled Drug Delivery, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Cornelus F. van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Twan Lammers
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
- Department of Experimental Molecular Imaging (ExMI), Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Clinic, Aachen, Germany
- Department of Controlled Drug Delivery, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
137
|
Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm 2015; 93:52-79. [PMID: 25813885 DOI: 10.1016/j.ejpb.2015.03.018] [Citation(s) in RCA: 1144] [Impact Index Per Article: 114.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 02/08/2023]
Abstract
Cancer is the second worldwide cause of death, exceeded only by cardiovascular diseases. It is characterized by uncontrolled cell proliferation and an absence of cell death that, except for hematological cancers, generates an abnormal cell mass or tumor. This primary tumor grows thanks to new vascularization and, in time, acquires metastatic potential and spreads to other body sites, which causes metastasis and finally death. Cancer is caused by damage or mutations in the genetic material of the cells due to environmental or inherited factors. While surgery and radiotherapy are the primary treatment used for local and non-metastatic cancers, anti-cancer drugs (chemotherapy, hormone and biological therapies) are the choice currently used in metastatic cancers. Chemotherapy is based on the inhibition of the division of rapidly growing cells, which is a characteristic of the cancerous cells, but unfortunately, it also affects normal cells with fast proliferation rates, such as the hair follicles, bone marrow and gastrointestinal tract cells, generating the characteristic side effects of chemotherapy. The indiscriminate destruction of normal cells, the toxicity of conventional chemotherapeutic drugs, as well as the development of multidrug resistance, support the need to find new effective targeted treatments based on the changes in the molecular biology of the tumor cells. These novel targeted therapies, of increasing interest as evidenced by FDA-approved targeted cancer drugs in recent years, block biologic transduction pathways and/or specific cancer proteins to induce the death of cancer cells by means of apoptosis and stimulation of the immune system, or specifically deliver chemotherapeutic agents to cancer cells, minimizing the undesirable side effects. Although targeted therapies can be achieved directly by altering specific cell signaling by means of monoclonal antibodies or small molecules inhibitors, this review focuses on indirect targeted approaches that mainly deliver chemotherapeutic agents to molecular targets overexpressed on the surface of tumor cells. In particular, we offer a detailed description of different cytotoxic drug carriers, such as liposomes, carbon nanotubes, dendrimers, polymeric micelles, polymeric conjugates and polymeric nanoparticles, in passive and active targeted cancer therapy, by enhancing the permeability and retention or by the functionalization of the surface of the carriers, respectively, emphasizing those that have received FDA approval or are part of the most important clinical studies up to date. These drug carriers not only transport the chemotherapeutic agents to tumors, avoiding normal tissues and reducing toxicity in the rest of the body, but also protect cytotoxic drugs from degradation, increase the half-life, payload and solubility of cytotoxic agents and reduce renal clearance. Despite the many advantages of all the anticancer drug carriers analyzed, only a few of them have reached the FDA approval, in particular, two polymer-protein conjugates, five liposomal formulations and one polymeric nanoparticle are available in the market, in contrast to the sixteen FDA approval of monoclonal antibodies. However, there are numerous clinical trials in progress of polymer-protein and polymer-drug conjugates, liposomal formulations, including immunoliposomes, polymeric micelles and polymeric nanoparticles. Regarding carbon nanotubes or dendrimers, there are no FDA approvals or clinical trials in process up to date due to their unresolved toxicity. Moreover, we analyze in detail the more promising and advanced preclinical studies of the particular case of polymeric nanoparticles as carriers of different cytotoxic agents to active and passive tumor targeting published in the last 5 years, since they have a huge potential in cancer therapy, being one of the most widely studied nano-platforms in this field in the last years. The interest that these formulations have recently achieved is stressed by the fact that 90% of the papers based on cancer therapeutics with polymeric nanoparticles have been published in the last 6 years (PubMed search).
Collapse
|
138
|
Polymeric micelles and nanoemulsions as tumor-targeted drug carriers: Insight through intravital imaging. J Control Release 2015; 206:153-60. [PMID: 25776738 DOI: 10.1016/j.jconrel.2015.03.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/04/2015] [Accepted: 03/09/2015] [Indexed: 11/21/2022]
Abstract
Intravital imaging of nanoparticle extravasation and tumor accumulation has revealed, for the first time, detailed features of carrier and drug behavior in circulation and tissue that suggest new directions for optimization of drug nanocarriers. Using intravital fluorescent microscopy, the extent of the extravasation, diffusion in the tissue, internalization by tissue cells, and uptake by the RES system were studied for polymeric micelles, nanoemulsions, and nanoemulsion-encapsulated drug. Discrimination of vascular and tissue compartments in the processes of micelle and nanodroplet extravasation and tissue accumulation was possible. A simple 1-D continuum model was suggested that allowed discriminating between various kinetic regimes of nanocarrier (or released drug) internalization in tumors of various sizes and cell density. The extravasation and tumor cell internalization occurred much faster for polymeric micelles than for nanoemulsion droplets. Fast micelle internalization resulted in the formation of a perivascular fluorescent coating around blood vessels. A new mechanism of micelle extravasation and internalization was suggested, based on the fast extravasation and internalization rates of copolymer unimers while maintaining micelle/unimer equilibrium in the circulation. The data suggested that to be therapeutically effective, nanoparticles with high internalization rate should manifest fast diffusion in the tumor tissue in order to avoid generation of concentration gradients that induce drug resistance. However an extra-fast diffusion should be avoided as it may result in the flow of extravasated nanoparticles from the tumor to normal organs, which would compromise targeting efficiency. The extravasation kinetics were different for nanodroplets and nanodroplet-encapsulated drug F-PTX suggesting a premature release of some fraction of the drug from the carrier. In conclusion, the development of an "ideal" drug carrier should involve the optimization of both drug retention and carrier diffusion parameters.
Collapse
|
139
|
He X, Li L, Su H, Zhou D, Song H, Wang L, Jiang X. Poly(ethylene glycol)-block-poly(ε-caprolactone)-and phospholipid-based stealth nanoparticles with enhanced therapeutic efficacy on murine breast cancer by improved intracellular drug delivery. Int J Nanomedicine 2015; 10:1791-804. [PMID: 25784805 PMCID: PMC4356685 DOI: 10.2147/ijn.s75186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Effective anticancer drug delivery to the tumor site without rapid body clearance is a prerequisite for successful chemotherapy. 1,2-distearoyl-sn-glycero-3-phospho-ethanolamine-N-(methoxy[polyethyleneglycol]-2000) (DSPE-PEG2000) has been widely used in the preparation of stealth liposomes. Although PEG chains can efficiently preserve liposomes from rapid clearance by the reticuloendothelial system (RES), its application has been hindered by poor cellular uptake and unsatisfactory therapeutic effect. METHODS To address the dilemma, we presented a facile approach to fabricate novel stealth nanoparticles generated by poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL), soybean phosphatidylcholine (SPC), and cholesterol, namely LPPs (L represented lipid and PP represented PEG-b-PCL), for the delivery of anticancer drug paclitaxel (PTX). LPPs were prepared using the thin film hydration method. Two PEG-b-PCL polymers with different molecular weights (MW; PEG2000-b-PCL2000, MW: 4,000 Da and PEG5000-b-PCL5000, MW: 10,000 Da) were used to fabricate stealth nanoparticles. Conventional PEGylated liposome (LDP2000, L represented lipid and DP2000 represented DSPE-PEG2000) composed of SPC, cholesterol, and DSPE-PEG2000 was used as the control. The physical properties, cellular uptake, endocytosis pathway, cytotoxicity, pharmacokinetics, tumor accumulation, and anticancer efficacy of free PTX, PTX-loaded LPPs, and LDP2000 were systemically investigated after injection into 4T1 breast tumor-bearing mice. RESULTS LPPs were vesicles around 100 nm in size with negative zeta potential. With enhanced stability, LPPs achieved sustainable release of cancer therapeutics. The cellular uptake level was closely related to the PEG chain length of PEG-b-PCL; a shorter PEG chain resulted in higher cellular uptake. Moreover, the cellular internalization of LPP2000 modified by PEG2000-b-PCL2000 on 4T1 cells was 2.1-fold higher than LDP2000 due to the improved stability of LPP2000. The cytotoxicity of PTX-loaded LPP2000 was also higher than that of LDP2000 and LPP5000 as observed using a WST-8 assay, while blank LPPs showed negligible toxicity. Consistent with the results of the in vitro study, in vivo experiments showed that LPPs allowed significantly improved bioavailability and prolonged T1/2β as compared to free PTX injection. More importantly, LPPs mainly accumulated at the tumor site, probably due to the enhanced permeation and retention effect (EPR effect). As a nanomedicine, LPP2000 (tumor inhibition rate of 75.1%) significantly enhanced the therapeutic effect of PTX in 4T1 breast tumor-bearing mice by inhibiting tumor growth compared to LDP2000 and LPP5000 (tumor inhibition rates of 56.3% and 49.5%, respectively). CONCLUSION Modification of liposomes with PEG2000-b-PCL2000 can simultaneously improve drug accumulation at the target tumor site and tumor cells, showing great promise for utilization as a PEG modification tool in the fabrication of stealth nanoparticles for cancer chemotherapy.
Collapse
Affiliation(s)
- Xiaodan He
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hong Su
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dinglun Zhou
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hongmei Song
- HitGen Ltd., Chengdu, Sichuan, People's Republic of China
| | - Ling Wang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xuehua Jiang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
140
|
Talelli M, Barz M, Rijcken CJ, Kiessling F, Hennink WE, Lammers T. Core-Crosslinked Polymeric Micelles: Principles, Preparation, Biomedical Applications and Clinical Translation. NANO TODAY 2015; 10:93-117. [PMID: 25893004 PMCID: PMC4398985 DOI: 10.1016/j.nantod.2015.01.005] [Citation(s) in RCA: 371] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Polymeric micelles (PM) are extensively used to improve the delivery of hydrophobic drugs. Many different PM have been designed and evaluated over the years, and some of them have steadily progressed through clinical trials. Increasing evidence suggests, however, that for prolonged circulation times and for efficient EPR-mediated drug targeting to tumors and to sites of inflammation, PM need to be stabilized, to prevent premature disintegration. Core-crosslinking is among the most popular methods to improve the in vivo stability of PM, and a number of core-crosslinked polymeric micelles (CCPM) have demonstrated promising efficacy in animal models. The latter is particularly true for CCPM in which (pro-) drugs are covalently entrapped. This ensures proper drug retention in the micelles during systemic circulation, efficient drug delivery to pathological sites via EPR, and tailorable drug release kinetics at the target site. We here summarize recent advances in the CCPM field, addressing the chemistry involved in preparing them, their in vitro and in vivo performance, potential biomedical applications, and guidelines for efficient clinical translation.
Collapse
Affiliation(s)
- Marina Talelli
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Immunology and Oncology and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)/CSIC, Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | | | - Fabian Kiessling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Twan Lammers
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
- Department of Controlled Drug Delivery, University of Twente and MIRA Institute for Biomedical Technology and Technical Medicine, Enschede, The Netherlands
| |
Collapse
|
141
|
Luk B, Zhang L. Current advances in polymer-based nanotheranostics for cancer treatment and diagnosis. ACS APPLIED MATERIALS & INTERFACES 2014; 6:21859-73. [PMID: 25014486 PMCID: PMC4278687 DOI: 10.1021/am5036225] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 07/11/2014] [Indexed: 05/05/2023]
Abstract
Nanotheranostics is a relatively new, fast-growing field that combines the advantages of treatment and diagnosis via a single nanoscale carrier. The ability to bundle both therapeutic and diagnostic capabilities into one package offers exciting prospects for the development of novel nanomedicine. Nanotheranostics can deliver treatment while simultaneously monitoring therapy response in real-time, thereby decreasing the potential of over- or under-dosing patients. Polymer-based nanomaterials, in particular, have been used extensively as carriers for both therapeutic and bioimaging agents and thus hold great promise for the construction of multifunctional theranostic formulations. Herein, we review recent advances in polymer-based systems for nanotheranostics, with a particular focus on their applications in cancer research. We summarize the use of polymer nanomaterials for drug delivery, gene delivery, and photodynamic therapy, combined with imaging agents for magnetic resonance imaging, radionuclide imaging, and fluorescence imaging.
Collapse
Affiliation(s)
- Brian
T. Luk
- Department
of NanoEngineering
and Moores Cancer Center, University of
California, San Diego, La Jolla, California 92093, United States
| | - Liangfang Zhang
- Department
of NanoEngineering
and Moores Cancer Center, University of
California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
142
|
Badrzadeh F, Rahmati-Yamchi M, Badrzadeh K, Valizadeh A, Zarghami N, Farkhani SM, Akbarzadeh A. Drug delivery and nanodetection in lung cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2014; 44:618-34. [DOI: 10.3109/21691401.2014.975237] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
143
|
Ke X, Ng VWL, Ono RJ, Chan JM, Krishnamurthy S, Wang Y, Hedrick JL, Yang YY. Role of non-covalent and covalent interactions in cargo loading capacity and stability of polymeric micelles. J Control Release 2014; 193:9-26. [DOI: 10.1016/j.jconrel.2014.06.061] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/10/2014] [Accepted: 06/24/2014] [Indexed: 10/25/2022]
|
144
|
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Engineered nanoparticles for drug delivery in cancer therapy. Angew Chem Int Ed Engl 2014; 53:12320-64. [PMID: 25294565 DOI: 10.1002/anie.201403036] [Citation(s) in RCA: 804] [Impact Index Per Article: 73.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Indexed: 12/18/2022]
Abstract
In medicine, nanotechnology has sparked a rapidly growing interest as it promises to solve a number of issues associated with conventional therapeutic agents, including their poor water solubility (at least, for most anticancer drugs), lack of targeting capability, nonspecific distribution, systemic toxicity, and low therapeutic index. Over the past several decades, remarkable progress has been made in the development and application of engineered nanoparticles to treat cancer more effectively. For example, therapeutic agents have been integrated with nanoparticles engineered with optimal sizes, shapes, and surface properties to increase their solubility, prolong their circulation half-life, improve their biodistribution, and reduce their immunogenicity. Nanoparticles and their payloads have also been favorably delivered into tumors by taking advantage of the pathophysiological conditions, such as the enhanced permeability and retention effect, and the spatial variations in the pH value. Additionally, targeting ligands (e.g., small organic molecules, peptides, antibodies, and nucleic acids) have been added to the surface of nanoparticles to specifically target cancerous cells through selective binding to the receptors overexpressed on their surface. Furthermore, it has been demonstrated that multiple types of therapeutic drugs and/or diagnostic agents (e.g., contrast agents) could be delivered through the same carrier to enable combination therapy with a potential to overcome multidrug resistance, and real-time readout on the treatment efficacy. It is anticipated that precisely engineered nanoparticles will emerge as the next-generation platform for cancer therapy and many other biomedical applications.
Collapse
Affiliation(s)
- Tianmeng Sun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA)
| | | | | | | | | | | |
Collapse
|
145
|
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Maßgeschneiderte Nanopartikel für den Wirkstofftransport in der Krebstherapie. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201403036] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
146
|
Polymeric nano-micelles: versatile platform for targeted delivery in cancer. Ther Deliv 2014; 5:1101-21. [DOI: 10.4155/tde.14.69] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Polymeric micelles are among the most promising delivery systems in nanomedicine. The growing interest in polymeric micelles as drug delivery vehicle is promoted by the advantages they offer for hydrophobic anticancer agents. The size of most polymeric micelles lies within the range 10–100 nm ensuring that they can selectively leave the circulation at tumor site via the enhanced permeability and retention effect. Their unique structure allows them to solubilize hydrophobic drugs, prolongs their circulatory half-life and eventually leads to enhanced therapeutic efficacy. In addition, they can undergo several structural modifications to further augment tumor cell uptake. In this review, we will discuss various micellar systems that have been studied in preclinical and clinical settings.
Collapse
|
147
|
Zhang Y, Huang Y, Zhao W, Lu J, Zhang P, Zhang X, Li J, Gao X, Venkataramanan R, Li S. Fmoc-conjugated PEG-vitamin E2 micelles for tumor-targeted delivery of paclitaxel: enhanced drug-carrier interaction and loading capacity. AAPS JOURNAL 2014; 16:1282-91. [PMID: 25193267 DOI: 10.1208/s12248-014-9651-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 07/22/2014] [Indexed: 11/30/2022]
Abstract
The purpose of this study is to develop an improved drug delivery system for enhanced paclitaxel (PTX) loading capacity and formulation stability based on PEG5K-(vitamin E)2 (PEG5K-VE2) system. PEG5K-(fluorenylmethoxycarbonyl)-(vitamin E)2 (PEG5K-FVE2) was synthesized using lysine as the scaffold. PTX-loaded PEG5K-FVE2 micelles were prepared and characterized. Fluorescence intensity of Fmoc in the micelles was measured as an indicator of drug-carrier interaction. Cytotoxicity of the micelle formulations was tested on various tumor cell lines. The therapeutic efficacy and toxicity of PTX-loaded micelles were investigated using a syngeneic mouse model of breast cancer (4T1.2). Our data suggest that the PEG5K-FVE2 micelles have a low CMC value of 4 μg/mL and small sizes (~60 nm). The PTX loading capacity of PEG5K-FVE2 micelles was much higher than that of PEG5K-VE2 micelles. The Fmoc/PTX physical interaction was clearly demonstrated by a fluorescence quenching assay. PTX-loaded PEG5K-FVE2 micelles exerted more potent cytotoxicity than free PTX or Taxol formulation in vitro. Finally, intravenous injection of PTX-loaded PEG5K-FVE2 micelles showed superior anticancer activity compared with PEG5K-VE2 formulation with minimal toxicity in a mouse model of breast cancer. In summary, incorporation of a drug-interactive motif (Fmoc) into PEG5K-VE2 micelles represents an effective strategy to improve the micelle formulation for the delivery of PTX.
Collapse
Affiliation(s)
- Yifei Zhang
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, 639 Salk Hall, Pittsburgh, PA, 15261, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Zhang Y, Huang Y, Li S. Polymeric micelles: nanocarriers for cancer-targeted drug delivery. AAPS PharmSciTech 2014; 15:862-71. [PMID: 24700296 DOI: 10.1208/s12249-014-0113-z] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/13/2014] [Indexed: 11/30/2022] Open
Abstract
Polymeric micelles represent an effective delivery system for poorly water-soluble anticancer drugs. With small size (10-100 nm) and hydrophilic shell of PEG, polymeric micelles exhibit prolonged circulation time in the blood and enhanced tumor accumulation. In this review, the importance of rational design was highlighted by summarizing the recent progress on the development of micellar formulations. Emphasis is placed on the new strategies to enhance the drug/carrier interaction for improved drug-loading capacity. In addition, the micelle-forming drug-polymer conjugates are also discussed which have both drug-loading function and antitumor activity.
Collapse
|
149
|
Antitumor activity of PEGylated biodegradable nanoparticles for sustained release of docetaxel in triple-negative breast cancer. Int J Pharm 2014; 473:55-63. [PMID: 24992317 DOI: 10.1016/j.ijpharm.2014.06.058] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 06/27/2014] [Accepted: 06/28/2014] [Indexed: 11/21/2022]
Abstract
With the aim to find novel therapeutical approaches for triple-negative breast cancer (TNBC) treatment, we have developed a powder for i.v. injection based on cyclodextrins and docetaxel (DTX)-loaded polyethyleneglycol-poly(epsilon-caprolactone) nanoparticles (DTX-NPs). Nanoparticles are designed to concentrate at tumor level by enhanced permeability and retention effect and release drug cargo at a sustained rate in the blood and in tumor interstitium. DTX-NPs of around 70 nm, shielding proteins and allowing a sustained DTX release for about 30 days, were produced by melting sonication technique. DTX-NPs were associated to hydroxypropyl-β-cyclodextrin to give a powder for injection with excellent dispersibility and suitable for i.v. administration. DTX-NPs were as efficient as free DTX in inhibiting cell growth of MDA-MB231 cells, even at low concentrations, and displayed a comparable in vivo antitumor efficacy and better survival in a TNBC animal model as compared with DTX commercial formulation (Taxotere(®)). In conclusion, PEGylated biodegradable DTX-NPs highlighted their potential in the treatment of aggressive TNBC providing a foundation for future clinical studies.
Collapse
|
150
|
Ahn HK, Jung M, Sym SJ, Shin DB, Kang SM, Kyung SY, Park JW, Jeong SH, Cho EK. A phase II trial of Cremorphor EL-free paclitaxel (Genexol-PM) and gemcitabine in patients with advanced non-small cell lung cancer. Cancer Chemother Pharmacol 2014; 74:277-82. [PMID: 24906423 PMCID: PMC4112044 DOI: 10.1007/s00280-014-2498-5] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 05/23/2014] [Indexed: 11/29/2022]
Abstract
Purpose Genexol-PM is a Cremorphor EL (CrEL)-free polymeric micelle formulation of paclitaxel that allows higher-dose administration with less hypersensitivity. This study was designed to evaluate the efficacy and safety of Genexol-PM and gemcitabine combination in advanced non-small cell lung cancer patients as a first-line treatment. Patients and methods This is a prospective, single-arm, single-center phase II study. Patients with advanced NSCLC received Genexol-PM at 230 mg/m2 on day 1 and gemcitabine 1,000 mg/m2 on day 1 and day 8 of a 3-week cycle. Six cycles of chemotherapy were planned unless there was disease progression. The primary endpoint was overall response rate. Results Forty-three patients received the study drugs with a median of 4 cycles per patient (range 1–6). The overall response rate was 46.5 %. The median progression-free survival was 4.0 months (95 % CI 2.0–6.0 months), and median overall survival was 14.8 months (95 % CI 9.1–20.5 months). The most common toxicities were anemia (n = 29, 67 %), asthenia (n = 17, 40 %), myalgia (n = 16, 37 %), peripheral neuropathy (n = 15, 35 %), and diarrhea (n = 12, 30 %). The most common grade 3/4 adverse events were neutropenia (n = 7, 16 %) and pneumonia (n = 5, 12 %). Two patients died of pneumonia and dyspnea. Conclusions CrEL-free paclitaxel in combination with gemcitabine demonstrated favorable antitumor activity with little emetogenicities in non-small cell lung cancer patients. However, frequent grade 3/4 toxicities were observed, and safety should be evaluated thoroughly in future studies.
Collapse
Affiliation(s)
- Hee Kyung Ahn
- Division of Hematology and Oncology, Department of Internal Medicine, Gachon University Gil Medical Center, 1198 Guwol-dong, Namdong-gu, Incheon, 405-760, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|