51
|
A Novel Gd-DTPA-conjugated Poly(L-γ-glutamyl-glutamine)-paclitaxel Polymeric Delivery System for Tumor Theranostics. Sci Rep 2017. [PMID: 28630436 PMCID: PMC5476566 DOI: 10.1038/s41598-017-03633-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The conventional chemotherapeutics could not be traced in vivo and provide timely feedback on the clinical effectiveness of drugs. In this study, poly(L-γ-glutamyl-glutamine)-paclitaxel (PGG-PTX), as a model polymer, was chemically conjugated with Gd-DTPA (Gd-diethylenetriaminepentaacetic acid), a T1-contrast agent of MRI, to prepare a Gd-DTPA-conjugated PGG-PTX (PGG-PTX-DTPA-Gd) delivery system used for tumor theranostics. PGG-PTX-DTPA-Gd can be self-assembled to NPs in water with a z-average hydrodynamic diameter about 35.9 nm. The 3 T MRI results confirmed that the relaxivity of PGG-PTX-DTPA-Gd NPs (r1 = 18.98 mM−1S−1) was increased nearly 4.9 times compared with that of free Gd-DTPA (r1 = 3.87 mM−1S−1). The in vivo fluorescence imaging results showed that PGG-PTX-DTPA-Gd NPs could be accumulated in the tumor tissue of NCI-H460 lung cancer animal model by EPR effect, which was similar to PGG-PTX NPs. The MRI results showed that compared with free Gd-DTPA, PGG-PTX-DTPA-Gd NPs showed significantly enhanced and prolonged signal intensity in tumor tissue, which should be attributed to the increased relaxivity and tumor accumulation. PGG-PTX-DTPA-Gd NPs also showed effective antitumor effect in vivo. These results indicated that PGG-PTX-DTPA-Gd NPs are an effective delivery system for tumor theranostics, and should have a potential value in personalized treatment of tumor.
Collapse
|
52
|
MacEwan SR, Chilkoti A. From Composition to Cure: A Systems Engineering Approach to Anticancer Drug Carriers. Angew Chem Int Ed Engl 2017; 56:6712-6733. [PMID: 28028871 PMCID: PMC6372097 DOI: 10.1002/anie.201610819] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Indexed: 12/21/2022]
Abstract
The molecular complexity and heterogeneity of cancer has led to a persistent, and as yet unsolved, challenge to develop cures for this disease. The pharmaceutical industry focuses the bulk of its efforts on the development of new drugs, but an alternative approach is to improve the delivery of existing drugs with drug carriers that can manipulate when, where, and how a drug exerts its therapeutic effect. For the treatment of solid tumors, systemically delivered drug carriers face significant challenges that are imposed by the pathophysiological barriers that lie between their site of administration and their site of therapeutic action in the tumor. Furthermore, drug carriers face additional challenges in their translation from preclinical validation to clinical approval and adoption. Addressing this diverse network of challenges requires a systems engineering approach for the rational design of optimized carriers that have a realistic prospect for translation from the laboratory to the patient.
Collapse
Affiliation(s)
- Sarah R MacEwan
- Department of Biomedical Engineering, Duke University, P.O. Box 90281, Durham, NC, 27708, USA
- Research Triangle MRSEC, Durham, NC, 27708, USA
- Present address: Institute for Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, P.O. Box 90281, Durham, NC, 27708, USA
- Research Triangle MRSEC, Durham, NC, 27708, USA
| |
Collapse
|
53
|
Design of Y-shaped targeting material for liposome-based multifunctional glioblastoma-targeted drug delivery. J Control Release 2017; 255:132-141. [DOI: 10.1016/j.jconrel.2017.04.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 03/27/2017] [Accepted: 04/05/2017] [Indexed: 12/29/2022]
|
54
|
Fu YN, Li Y, Li G, Yang L, Yuan Q, Tao L, Wang X. Adaptive Chitosan Hollow Microspheres as Efficient Drug Carrier. Biomacromolecules 2017; 18:2195-2204. [DOI: 10.1021/acs.biomac.7b00592] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ya-nan Fu
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, People’s Republic of China
| | - Yongsan Li
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, People’s Republic of China
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Guofeng Li
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, People’s Republic of China
| | - Lei Yang
- Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100021, People’s Republic of China
| | - Qipeng Yuan
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, People’s Republic of China
| | - Lei Tao
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Xing Wang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, People’s Republic of China
| |
Collapse
|
55
|
Belhadj Z, Zhan C, Ying M, Wei X, Xie C, Yan Z, Lu W. Multifunctional targeted liposomal drug delivery for efficient glioblastoma treatment. Oncotarget 2017; 8:66889-66900. [PMID: 28978003 PMCID: PMC5620143 DOI: 10.18632/oncotarget.17976] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/21/2017] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) has been considered to be the most malignant brain tumors. Due to the existence of various barriers including the blood–brain barrier (BBB) and blood–brain tumor barrier (BBTB) greatly hinder the accumulation and deep penetration of chemotherapeutics, the treatment of glioma remains to be the most challenging task in clinic. In order to circumvent these hurdles, we developed a multifunctional liposomal glioma-targeted drug delivery system (c(RGDyK)/pHA-LS) modified with cyclic RGD (c(RGDyK)) and p-hydroxybenzoic acid (pHA) in which c(RGDyK) could target integrin αvβ3 overexpressed on the BBTB and glioma cells and pHA could target dopamine receptors on the BBB. In vitro, c(RGDyK)/pHA-LS could target glioblastoma cells (U87), brain capillary endothelial cells (bEnd.3) and umbilical vein endothelial cells (HUVECs) through a comprehensive pathway. Besides, c(RGDyK)/pHA-LS could also increase the cytotoxicity of doxorubicin encapsulated in liposomes on glioblastoma cells, and was able to penetrate inside the glioma spheroids after traversing the in vitro BBB and BBTB. In vivo, we demonstrated the targeting ability of c(RGDyK)/pHA-LS to intracranial glioma. As expected, c(RGDyK)/pHA-LS/DOX showed a median survival time of 35 days, which was 2.31-, 1.76- and 1.5-fold higher than that of LS/DOX, c(RGDyK)-LS/DOX, and pHA-LS/DOX, respectively. The findings here suggested that the multifunctional glioma-targeted drug delivery system modified with both c(RGDyK) and pHA displayed strong antiglioma efficiency in vitro and in vivo, representing a promising platform for glioma therapy.
Collapse
Affiliation(s)
- Zakia Belhadj
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, P.R. China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Man Ying
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, P.R. China
| | - Xiaoli Wei
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, P.R. China.,State Key Laboratory of Medical Neurobiology & The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, P.R. China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, P.R. China
| | - Zhiqiang Yan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, P.R. China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, P.R. China.,State Key Laboratory of Medical Neurobiology & The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, P.R. China.,State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, P.R. China
| |
Collapse
|
56
|
Maisel K, Sasso MS, Potin L, Swartz MA. Exploiting lymphatic vessels for immunomodulation: Rationale, opportunities, and challenges. Adv Drug Deliv Rev 2017; 114:43-59. [PMID: 28694027 PMCID: PMC6026542 DOI: 10.1016/j.addr.2017.07.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/29/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
Lymphatic vessels are the primary route of communication from peripheral tissues to the immune system; as such, they represent an important component of local immunity. In addition to their transport functions, new immunomodulatory roles for lymphatic vessels and lymphatic endothelial cells have come to light in recent years, demonstrating that lymphatic vessels help shape immune responses in a variety of ways: promoting tolerance to self-antigens, archiving antigen for later presentation, dampening effector immune responses, and resolving inflammation, among others. In addition to these new biological insights, the growing field of immunoengineering has begun to explore therapeutic approaches to utilize or exploit the lymphatic system for immunotherapy.
Collapse
Affiliation(s)
- Katharina Maisel
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Maria Stella Sasso
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Lambert Potin
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA; École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Melody A Swartz
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA; Ben May Institute for Cancer Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
57
|
MacEwan SR, Chilkoti A. Von der Zusammensetzung zur Heilung: ein systemtechnischer Ansatz zur Entwicklung von Trägern für Tumortherapeutika. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201610819] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Sarah R. MacEwan
- Department of Biomedical Engineering; Duke University; P.O. Box 90281 Durham NC 27708 USA
- Research Triangle MRSEC; Durham NC 27708 USA
- Institute for Molecular Engineering; University of Chicago; Chicago IL 60637 USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering; Duke University; P.O. Box 90281 Durham NC 27708 USA
- Research Triangle MRSEC; Durham NC 27708 USA
| |
Collapse
|
58
|
Singh Y, Pawar VK, Meher JG, Raval K, Kumar A, Shrivastava R, Bhadauria S, Chourasia MK. Targeting tumor associated macrophages (TAMs) via nanocarriers. J Control Release 2017; 254:92-106. [DOI: 10.1016/j.jconrel.2017.03.395] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
|
59
|
Yu X, Li A, Zhao C, Yang K, Chen X, Li W. Ultrasmall Semimetal Nanoparticles of Bismuth for Dual-Modal Computed Tomography/Photoacoustic Imaging and Synergistic Thermoradiotherapy. ACS NANO 2017; 11:3990-4001. [PMID: 28395135 DOI: 10.1021/acsnano.7b00476] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Multifunctional nanomaterials with integrated diagnostic and therapeutic functions, combination therapy to enhance treatment efficacy, as well as low toxicity have drawn tremendous attentions. Herein, we report a multifunctional theranostic agent based on peptide (LyP-1)-labeled ultrasmall semimetal nanoparticles of bismuth (Bi-LyP-1 NPs). Ultrasmall Bi NPs (3.6 nm) were facilely synthesized using oleylamine as the reducing agent and exhibited a higher tumor accumulation after being conjugated with the tumor-homing peptide LyP-1. The abilities to absorb both ionizing radiation and the second near-infrared (NIR-II) window laser radiation ensured that Bi-LyP-1 NPs are capable of dual-modal computed tomography/photoacoustic imaging and efficient synergistic NIR-II photothermal/radiotherapy of tumors. Moreover, Bi-LyP-1 NPs could be rapidly cleared from mice through both renal and fecal clearance and almost completely cleared after 30 days. Such multifunctional nanoparticles as efficient cancer theranostic agents, coupled with fast clearance and low toxicity, shed light on the future use of semimetal nanoparticles for biomedicine.
Collapse
Affiliation(s)
- Xujiang Yu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Ang Li
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Chengzhi Zhao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Kai Yang
- School of Radiation Medicine and Protection (SRMP) and School of Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University , Suzhou, Jiangsu 215123, People's Republic of China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH) , Bethesda, Maryland 20892, United States
| | - Wanwan Li
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| |
Collapse
|
60
|
Qin H, Ding Y, Mujeeb A, Zhao Y, Nie G. Tumor Microenvironment Targeting and Responsive Peptide-Based Nanoformulations for Improved Tumor Therapy. Mol Pharmacol 2017; 92:219-231. [PMID: 28420679 DOI: 10.1124/mol.116.108084] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/12/2017] [Indexed: 12/20/2022] Open
Abstract
The tumor microenvironment participates in all stages of tumor progression and has emerged as a promising therapeutic target for cancer therapy. Rapid progress in the field of molecular self-assembly using various biologic molecules has resulted in the fabrication of nanoformulations that specifically target and regulate microenvironment components to inhibit tumor growth. This inhibition process is based on differentiating between biophysicochemical cues guiding tumor and normal tissue microenvironments. Peptides and peptide derivatives, owing to their biocompatibility, chemical versatility, bioactivity, environmental sensitivity, and biologic recognition abilities, have been widely used as building blocks to construct multifunctional nanostructures for targeted drug delivery and controlled release. Several groups of peptides have been identified as having the ability to penetrate plasma membranes, regulate the essential signaling pathways of angiogenesis and immune reactions, and recognize key components in the tumor microenvironment (such as vascular systems, stromal cells, and abnormal tumor biophysicochemical features). Thus, using different modules, various functional peptides, and their derivatives can be integrated into nanoformulations specifically targeting the tumor microenvironment with increased selectivity, on-demand response, elevated cellular uptake, and improved tumor therapy. In this review, we introduce several groups of functional peptides and highlight peptide-based nanoformulations that specifically target the tumor microenvironment. We also provide our perspective on the development of smart drug-delivery systems with enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (H.Q., Y D., A.M., Y. Z., G.N.), and University of Chinese Academy of Sciences (H.Q., Y.D., Y.Z., G.N.), Beijing, China
| | - Yanping Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (H.Q., Y D., A.M., Y. Z., G.N.), and University of Chinese Academy of Sciences (H.Q., Y.D., Y.Z., G.N.), Beijing, China
| | - Ayeesha Mujeeb
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (H.Q., Y D., A.M., Y. Z., G.N.), and University of Chinese Academy of Sciences (H.Q., Y.D., Y.Z., G.N.), Beijing, China
| | - Ying Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (H.Q., Y D., A.M., Y. Z., G.N.), and University of Chinese Academy of Sciences (H.Q., Y.D., Y.Z., G.N.), Beijing, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (H.Q., Y D., A.M., Y. Z., G.N.), and University of Chinese Academy of Sciences (H.Q., Y.D., Y.Z., G.N.), Beijing, China
| |
Collapse
|
61
|
Zhou JE, Yu J, Gao L, Sun L, Peng T, Wang J, Zhu J, Lu W, Zhang L, Yan Z, Yu L. iNGR-Modified Liposomes for Tumor Vascular Targeting and Tumor Tissue Penetrating Delivery in the Treatment of Glioblastoma. Mol Pharm 2017; 14:1811-1820. [DOI: 10.1021/acs.molpharmaceut.7b00101] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jing-e Zhou
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| | - Jing Yu
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| | - Lipeng Gao
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| | - Lei Sun
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| | - Ting Peng
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| | - Jing Wang
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| | - Jianzhong Zhu
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai 201203, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People’s
Hospital, Shaoxing Hospital of ZheJiang University, Shaoxing 312000 People’s Republic of China
| | - Zhiqiang Yan
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| | - Lei Yu
- Institute of Biomedical
Engineering, Technology, Shanghai Engineering Research Center of Molecular
Therapeutics, New Drug Development, School of Chemistry, Molecular
Engineering, East China Normal University, Shanghai 200062, China
| |
Collapse
|
62
|
Kuang H, Ku SH, Kokkoli E. The design of peptide-amphiphiles as functional ligands for liposomal anticancer drug and gene delivery. Adv Drug Deliv Rev 2017; 110-111:80-101. [PMID: 27539561 DOI: 10.1016/j.addr.2016.08.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/12/2016] [Accepted: 08/05/2016] [Indexed: 12/25/2022]
Abstract
Liposomal nanomedicine has led to clinically useful cancer therapeutics like Doxil and DaunoXome. In addition, peptide-functionalized liposomes represent an effective drug and gene delivery vehicle with increased cancer cell specificity, enhanced tumor-penetrating ability and high tumor growth inhibition. The goal of this article is to review the recently published literature of the peptide-amphiphiles that were used to functionalize liposomes, to highlight successful designs that improved drug and gene delivery to cancer cells in vitro, and cancer tumors in vivo, and to discuss the current challenges of designing these peptide-decorated liposomes for effective cancer treatment.
Collapse
|
63
|
Ying M, Zhan C, Wang S, Yao B, Hu X, Song X, Zhang M, Wei X, Xiong Y, Lu W. Liposome-Based Systemic Glioma-Targeted Drug Delivery Enabled by All-d Peptides. ACS APPLIED MATERIALS & INTERFACES 2016; 8:29977-29985. [PMID: 27797175 DOI: 10.1021/acsami.6b10146] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
As the most aggressive brain tumor, chemotherapy of malignant glioma remains to be extremely challenging in clinic. The blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) are physiological and pathological barriers preventing therapeutic drugs from reaching the glioma region. In addition, vasculogenic mimicry (VM) formed by invasive glioma cells instead of endothelial cells and angiogenesis are very common in glioma, leading to the poor prognosis and recurrence of glioma. An ideal drug delivery system for glioma chemotherapy needs to traverse the BBB and BBTB and then target VM, angiogenesis, and glioma cells. Herein we developed a liposome-based drug delivery system with the modification of proteolytically stable d-peptide ligands (dCDX/dA7R-LS). dCDX is a d-peptide ligand of nicotine acetylcholine receptors (nAChRs) capable of circumventing the BBB, and dA7R is a d-peptide ligand of vascular endothelial growth factor receptor 2 (VEGFR2) and neuropilin-1 (NRP-1) overexpressed on angiogenesis, VM, and glioma, presenting excellent glioma-homing property. dCDX/dA7R-LS could efficiently internalize into the brain capillary endothelial cells, glioma cells, tumor neovascular endothelial cells, and tumor spheroids and cross the in vitro BBB and BBTB models. Ex vivo imaging and in vivo immunofluorescence assays confirmed the superiority of dCDX/dA7R-LS in targeting intracranial glioma in comparison to plain liposomes or liposomes modified with an individual d-peptide ligand (either dCDX or dA7R). When loaded with doxorubicin, dCDX/dA7R-LS achieved the best antiglioma, antiangiogenesis, and anti-VM effects among all tested formulations. These results suggested that systemic glioma-targeted drug delivery enabled by all-d peptide ligands was promising for the antiglioma therapy.
Collapse
Affiliation(s)
- Man Ying
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Changyou Zhan
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University , Shanghai 200032, China
| | - Songli Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Bingxin Yao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Xuefeng Hu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Xianfei Song
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Mingfei Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Xiaoli Wei
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, The Collaborative Innovation Center for Brain Science, Fudan University , Shanghai 200032, China
| | - Yan Xiong
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education , Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, The Collaborative Innovation Center for Brain Science, Fudan University , Shanghai 200032, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University , Shanghai 200433, China
| |
Collapse
|
64
|
Ying M, Shen Q, Zhan C, Wei X, Gao J, Xie C, Yao B, Lu W. A stabilized peptide ligand for multifunctional glioma targeted drug delivery. J Control Release 2016; 243:86-98. [PMID: 27693752 DOI: 10.1016/j.jconrel.2016.09.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 09/15/2016] [Accepted: 09/28/2016] [Indexed: 12/18/2022]
Abstract
Peptide ligands consisting of l-amino acids are subject to proteolysis in vivo. When modified on the surface of nanocarriers, those peptide ligands would readily degrade and the targeting efficacy is significantly attenuated. It has received increasing scrutiny to design stable peptide ligands for targeted drug delivery. Here, we present the design of a stable peptide ligand by the formation of a head-to-tail amide bond as an example. Even though the linear l-peptide A7R (termed LA7R) can bind specifically to vascular endothelial growth factor receptor 2 (VEGFR2) and neuropilin-1 (NRP-1) that are overexpressed on glioma cells, neovasculature and glioma vasculogenic mimicry (VM), the tumor-homing capacity of LA7R is greatly impaired in vivo due to proteolysis (e.g. in the serum). A cyclic A7R (cA7R) peptide was identified by computer-aided peptide design and synthesized with high yield by combining solid phase peptide synthesis and native chemical ligation. The binding of cA7R to both receptors was theoretically and experimentally assessed. In our simulated model hydrophobic and ionic interactions dominated the binding of LA7R to receptors. It is very interesting that cA7R adopting a different structure from LA7R retained high binding affinities to receptors without affecting the hydrophobic and ionic interactions. After head-to-tail cyclization by the formation of an amide bond, cA7R exhibited exceptional stability in mouse serum. Either cA7R or LA7R was conjugated on the surface of doxorubicin (DOX) loaded liposomes (cA7R-LS/DOX or LA7R-LS/DOX). The results of in vitro cellular assays indicated that cA7R-LS/DOX not only displayed stronger anti-proliferative effect against glioma cells, but also demonstrated to be more efficient in destruction of VM and HUVEC tubes in comparison to LA7R-LS/DOX and plain liposomes (LS/DOX, without peptide conjugation). cA7R conjugation could achieve significantly higher accumulation of liposomes in glioma than did LA7R conjugation, which in turn, cA7R-LS/DOX could substantially suppress subcutaneous tumor growth when compared with other DOX formulations (free DOX, LS/DOX and LA7R-LS/DOX). The designed cyclic A7R exhibited the capability of targeting glioma cells, neovasculature and VM simultaneously in vivo. Considering the ease of synthesis, high binding affinity to receptors and increased stability of cA7R peptide in the present study, the design of head-to-tail cyclized peptides by the formation of amide bond based on computer-aided peptide design presents an alternative method to identify proteolytically stable peptide ligands.
Collapse
Affiliation(s)
- Man Ying
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Qing Shen
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Changyou Zhan
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China; Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoli Wei
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology, The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jie Gao
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Bingxin Yao
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China; State Key Laboratory of Medical Neurobiology, The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
65
|
Development of a multi-target peptide for potentiating chemotherapy by modulating tumor microenvironment. Biomaterials 2016; 108:44-56. [PMID: 27619239 DOI: 10.1016/j.biomaterials.2016.09.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/24/2016] [Accepted: 09/02/2016] [Indexed: 12/17/2022]
Abstract
Finding effective cures against aggressive malignancy remains a major challenge in cancer chemotherapy. Here, we report a "tadpole"-like peptide by covalently conjugating the alanine-alanine-asparagine "tail" residual to the cyclic tumor homing peptide iRGD (CCRGDKGPDC) to afford nRGD, which significantly enhanced tumoricidal effects of doxorubicin, by either co-administered as a physical mixture or as a targeting ligand covalently conjugated to the liposomal carrier. Given twice at an equivalent dose of 5 mg/kg, doxorubicin loaded liposomes modified with nRGD (nRGD-Lipo-Dox) showed excellent antitumor efficacy in 4T1 breast cancer mice, of which 44.4% remained alive for over 90 days without recurrence during the period of investigation. The dramatic improvement in antitumor efficacy was attributed to nRGD-Lipo-Dox which appeared to specifically interact with tumor vascular endothelial cells to achieve efficient tumor penetration, and modulate tumor microenvironment with depletion of tumor associated macrophages.
Collapse
|
66
|
Sucrose ester based cationic liposomes as effective non-viral gene vectors for gene delivery. Colloids Surf B Biointerfaces 2016; 145:454-461. [DOI: 10.1016/j.colsurfb.2016.05.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/16/2016] [Accepted: 05/11/2016] [Indexed: 01/01/2023]
|
67
|
Ying M, Shen Q, Liu Y, Yan Z, Wei X, Zhan C, Gao J, Xie C, Yao B, Lu W. Stabilized Heptapeptide A7R for Enhanced Multifunctional Liposome-Based Tumor-Targeted Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2016; 8:13232-13241. [PMID: 27195531 DOI: 10.1021/acsami.6b01300] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
(L)A7R (ATWLPPR) is a heptapeptide with high binding affinity in vitro to vascular endothelial growth factor receptor 2 (VEGFR2) and neuropilin-1 (NRP-1) overexpressed on glioma, glioma vasculogenic mimicry and neovasculature. However, its tumor targeting efficacy is significantly reduced in vivo due to proteolysis in blood circulation. To improve the in vivo stability and targeting efficacy, the retro inverso isomer of (L)A7R ((D)A7R) was developed for glioma-targeted drug delivery. (D)A7R was expected to have a similar binding affinity to its receptors in vitro (VEGFR2 and NRP-1), which was experimentally confirmed. In vivo, (D)A7R-modified liposomes achieved improved glioma-targeted efficiency than did (L)A7R-modified liposomes. After loading a chemotherapeutic agent (doxorubicin), (D)A7R-modified liposomes significantly inhibited subcutaneous model tumor in comparison to free doxorubicin, plain liposomes and (L)A7R-modified liposomes. In summary, the present study presented the potential of a proteolytically stable d-peptide ligand for in vivo tumor-targeted drug delivery.
Collapse
Affiliation(s)
- Man Ying
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Qing Shen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University , Shanghai 200433, China
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Zhiqiang Yan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of chemistry and molecular engineering, East China Normal University , Shanghai 200062, China
| | - Xiaoli Wei
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, The Collaborative Innovation Center for Brain Science, Fudan University , Shanghai 200032, China
| | - Changyou Zhan
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University , Shanghai 200032, China
| | - Jie Gao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Bingxin Yao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, (Fudan University), Ministry of Education , Shanghai 201203, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University , Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology, The Collaborative Innovation Center for Brain Science, Fudan University , Shanghai 200032, China
| |
Collapse
|
68
|
|
69
|
Paasonen L, Sharma S, Braun GB, Kotamraju VR, Chung TDY, She ZG, Sugahara KN, Yliperttula M, Wu B, Pellecchia M, Ruoslahti E, Teesalu T. New p32/gC1qR Ligands for Targeted Tumor Drug Delivery. Chembiochem 2016; 17:570-5. [PMID: 26895508 DOI: 10.1002/cbic.201500564] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Indexed: 11/08/2022]
Abstract
Cell surface p32, the target of LyP-1 homing peptide, is upregulated in tumors and atherosclerotic plaques and has been widely used as a receptor for systemic delivery of payloads. Here, we identified an improved LyP-1-mimicking peptide (TT1, CKRGARSTC). We used this peptide in a fluorescence polarization-based high-throughput screening of a 50,000-compound chemical library and identified a panel of compounds that bind p32 with low micromolar affinity. Among the hits identified in the screen, two compounds were shown to specifically bind to p32 in multiple assays. One of these compounds was chosen for an in vivo study. Nanoparticles surface-functionalized with this compound specifically adhered to surfaces coated with recombinant p32 and, when injected intravenously, homed to p32-expressing breast tumors in mice. This compound provides a lead for the development of p32-targeted affinity ligands that circumvent some of the limitations of peptide-based probes in guided drug delivery.
Collapse
Affiliation(s)
- Lauri Paasonen
- University of Helsinki, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, Viikinkaari 5E, Helsinki, 00014, Finland.,Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA
| | - Shweta Sharma
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA
| | - Gary B Braun
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA
| | - Venkata Ramana Kotamraju
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA.,Sanford Burnham Prebys Medical Discovery Institute Center for Nanomedicine, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Thomas D Y Chung
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA
| | - Zhi-Gang She
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA
| | - Kazuki N Sugahara
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA.,Department of Surgery, Columbia University College of Physicians and Surgeons, 1130 St Nicholas Avenue, Suite 217C, New York, NY, 100032, USA
| | - Marjo Yliperttula
- University of Helsinki, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, Viikinkaari 5E, Helsinki, 00014, Finland
| | - Bainan Wu
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA
| | - Maurizio Pellecchia
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA.,School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Erkki Ruoslahti
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA.,Sanford Burnham Prebys Medical Discovery Institute Center for Nanomedicine, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Tambet Teesalu
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA, 93027, USA. .,Sanford Burnham Prebys Medical Discovery Institute Center for Nanomedicine, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA. .,Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, Tartu, 50411, Estonia.
| |
Collapse
|
70
|
Wang Y, Wu B, Yang C, Liu M, Sum TC, Yong KT. Synthesis and Characterization of Mn:ZnSe/ZnS/ZnMnS Sandwiched QDs for Multimodal Imaging and Theranostic Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:534-546. [PMID: 26663023 DOI: 10.1002/smll.201503352] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Indexed: 06/05/2023]
Abstract
In this work, a facile aqueous synthesis method is optimized to produce Mn:ZnSe/ZnS/ZnMnS sandwiched quantum dots (SQDs). In this core-shell co-doped system, paramagnetic Mn(2+) ions are introduced as core and shell dopants to generate Mn phosphorescence and enhance the magnetic resonance imaging signal, respectively. T1 relaxivity of the nanoparticles can be improved and manipulated by raising the shell doping level. Steady state and time-resolved optical measurements suggest that, after high level shell doping, Mn phosphorescence of the core can be sustained by the sandwiched ZnS shell. Because the SQDs are free of toxic heavy metal compositions, excellent biocompatibility of the prepared nanocrystals is verified by in vitro MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. To explore the theranostic applications of SQDs, liposome-SQD assemblies are prepared and used for ex vivo optical and magnetic resonance imaging. In addition, these engineered SQDs as nanocarrier for gene delivery in therapy of Panc-1 cancer cells are employed. The therapeutic effects of the nanocrystals formulation are confirmed by gene expression analysis and cell viability assay.
Collapse
Affiliation(s)
- Yucheng Wang
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Bo Wu
- School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Chengbin Yang
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Maixian Liu
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Tze Chien Sum
- School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Ken-Tye Yong
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| |
Collapse
|
71
|
Hwang JY, Li Z, Loh XJ. Small molecule therapeutic-loaded liposomes as therapeutic carriers: from development to clinical applications. RSC Adv 2016. [DOI: 10.1039/c6ra09854a] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In this review, various methods and mechanisms for encapsulation of small therapeutic molecules in liposomes for targeted delivery and triggered release, as well as their potential in the clinical uses, are discussed.
Collapse
Affiliation(s)
- Jae Yoon Hwang
- Department of Materials Science and Engineering
- National University of Singapore
- Singapore 117576
- Singapore
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE)
- Singapore 117602
- Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE)
- Singapore 117602
- Singapore
- Department of Materials Science and Engineering
- National University of Singapore
| |
Collapse
|
72
|
Ganapathy V, Moghe PV, Roth CM. Targeting tumor metastases: Drug delivery mechanisms and technologies. J Control Release 2015; 219:215-223. [PMID: 26409123 PMCID: PMC4745901 DOI: 10.1016/j.jconrel.2015.09.042] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/22/2015] [Accepted: 09/22/2015] [Indexed: 12/17/2022]
Abstract
Primary sites of tumor are the focal triggers of cancers, yet it is the subsequent metastasis events that cause the majority of the morbidity and mortality. Metastatic tumor cells exhibit a phenotype that differs from that of the parent cells, as they represent a resistant, invasive subpopulation of the original tumor, may have acquired additional genetic or epigenetic alterations under exposure to prior chemotherapeutic or radiotherapeutic treatments, and reside in a microenvironment differing from that of its origin. This combination of resistant phenotype and distal location make tracking and treating metastases particularly challenging. In this review, we highlight some of the unique biological traits of metastasis, which in turn, inspire emerging strategies for targeted imaging of metastasized tumors and metastasis-directed delivery of therapeutics.
Collapse
Affiliation(s)
- Vidya Ganapathy
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA; Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, USA
| | - Charles M Roth
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA; Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, USA.
| |
Collapse
|
73
|
Wang B, Lv L, Wang Z, Jiang Y, Lv W, Liu X, Wang Z, Zhao Y, Xin H, Xu Q. Improved anti-glioblastoma efficacy by IL-13Rα2 mediated copolymer nanoparticles loaded with paclitaxel. Sci Rep 2015; 5:16589. [PMID: 26567528 PMCID: PMC4645113 DOI: 10.1038/srep16589] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/16/2015] [Indexed: 12/22/2022] Open
Abstract
Glioma presents one of the most malignant brain tumors, and the therapeutic effect is often limited due to the existence of brain tumor barrier. Based on interleukin-13 receptor α2 (IL-13Rα2) over-expression on glioma cell, it was demonstrated to be a potential receptor for glioma targeting. In this study, Pep-1-conjugated PEGylated nanoparticles loaded with paclitaxel (Pep-NP-PTX) were developed as a targeting drug delivery system for glioma treatment. The Pep-NP-PTX presented satisfactory size of 95.78 nm with narrow size distribution. Compared with NP-PTX, Pep-NP-PTX exhibited significantly enhanced cellular uptake in C6 cells (p < 0.001). The in vitro anti-proliferation evaluation showed that the IC50 were 146 ng/ml and 349 ng/ml of Pep-NP-PTX and NP-PTX, respectively. The in vivo fluorescent image results indicated that Pep-NP had higher specificity and efficiency in intracranial tumor accumulation. Following intravenous administration, Pep-NP-PTX could enhance the distribution of PTX in vivo glioma section, 1.98, 1.91 and 1.53-fold over that of NP-PTX group after 0.5, 1 and 4 h, respectively. Pep-NP-PTX could improve the anti-glioma efficacy with a median survival time of 32 days, which was significantly longer than that of PTX-NP (23 days) and Taxol(®) (22 days). In conclusion, Pep-NP-PTX is a potential targeting drug delivery system for glioma treatment.
Collapse
Affiliation(s)
- Baoyan Wang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.,Nanjing Drum Tower Hospital. The Affiliated Hospital of Nanjing University Medical School. Nanjing 210008, China
| | - Lingyan Lv
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zhi Wang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yan Jiang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wei Lv
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xin Liu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zhongyuan Wang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yue Zhao
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hongliang Xin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qunwei Xu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
74
|
Trevaskis NL, Kaminskas LM, Porter CJH. From sewer to saviour — targeting the lymphatic system to promote drug exposure and activity. Nat Rev Drug Discov 2015; 14:781-803. [DOI: 10.1038/nrd4608] [Citation(s) in RCA: 378] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
75
|
Torres Andón F, Alonso MJ. Nanomedicine and cancer immunotherapy – targeting immunosuppressive cells. J Drug Target 2015; 23:656-71. [DOI: 10.3109/1061186x.2015.1073295] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
76
|
Liposome-based glioma targeted drug delivery enabled by stable peptide ligands. J Control Release 2015; 218:13-21. [PMID: 26428462 DOI: 10.1016/j.jconrel.2015.09.059] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 07/07/2015] [Accepted: 09/28/2015] [Indexed: 01/05/2023]
Abstract
The treatment of glioma is one of the most challenging tasks in clinic. As an intracranial tumor, glioma exhibits many distinctive characteristics from other tumors. In particular, various barriers including enzymatic barriers in the blood and brain capillary endothelial cells, blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) rigorously prevent drug and drug delivery systems from reaching the tumor site. To tackle this dilemma, we developed a liposomal formulation to circumvent multiple-barriers by modifying the liposome surface with proteolytically stable peptides, (D)CDX and c(RGDyK). (D)CDX is a D-peptide ligand of nicotine acetylcholine receptors (nAChRs) on the BBB, and c(RGDyK) is a ligand of integrin highly expressed on the BBTB and glioma cells. Lysosomal compartments of brain capillary endothelial cells are implicated in the transcytosis of those liposomes. However, both peptide ligands displayed exceptional stability in lysosomal homogenate, ensuring that intact ligands could exert subsequent exocytosis from brain capillary endothelial cells and glioma targeting. In the cellular uptake studies, dually labeled liposomes could target both brain capillary endothelial cells and tumor cells, effectively traversing the BBB and BBTB monolayers, overcoming enzymatic barrier and targeting three-dimensional tumor spheroids. Its targeting ability to intracranial glioma was further verified in vivo by ex vivo imaging and histological studies. As a result, doxorubicin liposomes modified with both (D)CDX and c(RGDyK) presented better anti-glioma effect with prolonged median survival of nude mice bearing glioma than did unmodified liposomes and liposomes modified with individual peptide ligand. In conclusion, the liposome suggested in the present study could effectively overcome multi-barriers and accomplish glioma targeted drug delivery, validating its potential value in improving the therapeutic efficacy of doxorubicin for glioma.
Collapse
|
77
|
Abstract
The treatment for ischemic stroke is one of the most challenging problems and the therapeutic effect remains unsatisfied due to the poor permeation of drugs across the blood brain barrier (BBB). In this study, HAIYPRH (T7), a peptide that targeted to transferrin receptor (TfR) can mediate the transport of nanocarriers across the BBB, was conjugated to liposomes for ischemic stroke targeting treatment of a novel neuroprotectant (ZL006). T7-conjugated PEGylated liposomes (T7-P-LPs) loaded with ZL006 (T7-P-LPs/ZL006) were showed satisfactory vesicle size and size distribution. Furthermore, the cellular uptake results showed that T7 modification increased liposomes uptake by the brain capillary endothelial cells (BCECs) and little cytotoxicity of liposomes with or without ZL006 was observed. The in vivo biodistribution and near-infrared fluorescence imaging evidenced that T7 modification rendered liposomes significantly enhanced the transport of liposomes across the BBB. The pharmacodynamic study suggested that, T7-P-LPs/ZL006 exhibited reduced infarct volume and ameliorated neurological deficit compared with unmodified liposomes or free ZL006. T7-P-LPs/ZL006 could be targeted to brain and displayed remarkable neuroprotective effects. They could be used as a potential targeted drug delivery system of ischemic stroke treatment.
Collapse
|
78
|
Sun L, Wu Q, Peng F, Liu L, Gong C. Strategies of polymeric nanoparticles for enhanced internalization in cancer therapy. Colloids Surf B Biointerfaces 2015; 135:56-72. [PMID: 26241917 DOI: 10.1016/j.colsurfb.2015.07.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 06/19/2015] [Accepted: 07/07/2015] [Indexed: 02/05/2023]
Abstract
In order to achieve long circulation time and high drug accumulation in the tumor sites via the EPR effects, anticancer drugs have to be protected by non-fouling polymers such as poly(ethylene glycol) (PEG), poly(ethylene oxide) (PEO), dextran, and poly(acrylic acid) (PAA). However, the dense layer of stealth polymer also prohibits efficient uptake of anticancer drugs by target cancer cells. For cancer therapy, it is often more desirable to accomplish rapid cellular uptake after anticancer drugs arriving at the pathological site, which could on one hand maximize the therapeutic efficacy and on the other hand reduce probability of drug resistance in cells. In this review, special attention will be focused on the recent potential strategies that can enable drug-loaded polymeric nanoparticles to rapidly recognize cancer cells, leading to enhanced internalization.
Collapse
Affiliation(s)
- Lu Sun
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qinjie Wu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Feng Peng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lei Liu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Changyang Gong
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
79
|
Affiliation(s)
- Bhushan S Pattni
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | - Vladimir V Chupin
- Laboratory for Advanced Studies of Membrane Proteins, Moscow Institute of Physics and Technology , Dolgoprudny 141700, Russia
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States.,Department of Biochemistry, Faculty of Science, King Abdulaziz University , Jeddah 21589, Saudi Arabia
| |
Collapse
|
80
|
Ye T, Jiang X, Li J, Yang R, Mao Y, Li K, Li L, Chen F, Yao J, Wu Y, Yang X, Wang S, Pan W. Low molecular weight heparin mediating targeting of lymph node metastasis based on nanoliposome and enzyme–substrate interaction. Carbohydr Polym 2015; 122:26-38. [DOI: 10.1016/j.carbpol.2014.12.061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/17/2014] [Accepted: 12/28/2014] [Indexed: 01/26/2023]
|
81
|
Li X, Dong Q, Yan Z, Lu W, Feng L, Xie C, Xie Z, Su B, Liu M. MPEG-DSPE polymeric micelle for translymphatic chemotherapy of lymph node metastasis. Int J Pharm 2015; 487:8-16. [PMID: 25841567 DOI: 10.1016/j.ijpharm.2015.03.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/06/2015] [Accepted: 03/30/2015] [Indexed: 01/09/2023]
Abstract
Lymph node metastasis is one of the major pathways for tumor formation and it is difficult to deliver chemotherapeutics at therapeutic concentrations to lymph node metastasis. This study prepared methyl poly(ethylene glycol)-distearoylphosphatidylethanolamine/doxorubicin (MPEG-DSPE/DOX) micelle for the treatment of lymph node metastasis. The MPEG-DSPE/DOX micelle prepared were of spherical morphology with a particle size of 20 ± 5 nm. The uptake rates of DOX and MPEG-DSPE/DOX micelle by A375 cells were 51.2% and 88.7%, respectively. The phagocytosis rate of MPEG-DSPE/Rhodamine B micelle by RAW264.7 cells was 17.2-fold lower than for Rhodamine B alone. After subcutaneous injection, MPEG-DSPE micelle underwent lymphatic absorption and accumulated in popliteal lymph nodes. MPEG-DSPE/DOX micelle significantly alleviated damage to the subcutaneous tissue of the injection sites compared with DOX alone. We established a model of nude mice bearing lymph node metastasis of A375 cells. After subcutaneous injection, the weights of both the popliteal and iliac lymph nodes of the MPEG-DSPE/DOX micelle group were significantly lower than in the saline and DOX groups. MPEG-DSPE/DOX micelle effectively killed the tumor cells in popliteal and iliac lymph nodes. In conclusion, MPEG-DSPE micelle is a promising drug delivery system for the treatment of lymph node metastasis.
Collapse
Affiliation(s)
- Xue Li
- Key Laboratory of Smart Drug Delivery, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Qing Dong
- Key Laboratory of Smart Drug Delivery, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Zhiqiang Yan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Institutes for Advanced Interdisciplinary Research, East China Normal University, Shanghai 200062, China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Lingling Feng
- National Population and Family Planning Key Laboratory of Contraceptive Drugs and Devices, Shanghai Institute of Planned Parenthood Research, Shanghai 200032, China
| | - Cao Xie
- Key Laboratory of Smart Drug Delivery, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Zuoxu Xie
- Key Laboratory of Smart Drug Delivery, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Bingxia Su
- Key Laboratory of Smart Drug Delivery, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Min Liu
- Key Laboratory of Smart Drug Delivery, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| |
Collapse
|
82
|
Zhang Y, Deng C, Liu S, Wu J, Chen Z, Li C, Lu W. Active Targeting of Tumors through Conformational Epitope Imprinting. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201412114] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
83
|
Zhang Y, Deng C, Liu S, Wu J, Chen Z, Li C, Lu W. Active Targeting of Tumors through Conformational Epitope Imprinting. Angew Chem Int Ed Engl 2015; 54:5157-60. [DOI: 10.1002/anie.201412114] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Indexed: 12/18/2022]
|
84
|
Ye T, Xu W, Shi T, Yang R, Yang X, Wang S, Pan W. Targeted delivery of docetaxel to the metastatic lymph nodes: A comparison study between nanoliposomes and activated carbon nanoparticles. Asian J Pharm Sci 2015. [DOI: 10.1016/j.ajps.2014.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
85
|
Zeng K, Li J, Zhang Z, Yan M, Liao Y, Zhang X, Zhao C. Lipid-coated ZnO nanoparticles as lymphatic-targeted drug carriers: study on cell-specific toxicity in vitro and lymphatic targeting in vivo. J Mater Chem B 2015; 3:5249-5260. [DOI: 10.1039/c5tb00486a] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Lipid coated ZnO nanoparticles (LZnO NPs) were developed as a novel lymphatic drug delivery system. High lymphotropism and tumour cells selective toxicity ensure the nanoparticles being a promising candidate for treatment of tumor metastasis.
Collapse
Affiliation(s)
- Ke Zeng
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Jin Li
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Zhaoguo Zhang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Mina Yan
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Yunhui Liao
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Xuefei Zhang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| |
Collapse
|
86
|
Yu Z, Yu B, Kaye JB, Tang C, Chen S, Dong C, Shen B. Perspectives and Challenges of Cell-Penetrating Peptides in Effective siRNA Delivery. ACTA ACUST UNITED AC 2014. [DOI: 10.1142/s1793984414410165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Over the last two decades, hundreds of cell penetrating peptides (CPPs) have been intensively developed as drug and nucleic acid delivery vectors. In many cases, however, the efficient delivery of exogenous bioactive molecules through the plasma membrane to their targets remains a tremendous challenging issue. CPPs have attracted tremendous research interest as efficient cellular delivery vehicles due to their intrinsic ability to enter cells and mediate uptake of a wide range of macromolecular cargos, such as proteins, peptides, nucleic acids, drugs and nanoparticle carriers. This review presents and discusses the current perspectives of CPP-mediated siRNA delivery system. We focus on the CPP-mediated siRNA delivery approaches, and particular emphasis is placed on the strategies for the advantages and disadvantages for each delivery approach. Lastly, the cellular uptake mechanisms of CPPs and the specific challenges associated with each delivery system of siRNAs are discussed.
Collapse
Affiliation(s)
- Zhiqiang Yu
- Department of Physiology, Anhui Medical University, Hefei, Anhui 230032, P. R. China
- Center for BioEnergetics, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Bin Yu
- School of Pharmaceutical Sciences and New Drug Research & Development Center Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Justin Boy Kaye
- Center for BioEnergetics, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Chenhong Tang
- Center for BioEnergetics, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Shengxi Chen
- Center for BioEnergetics, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Chenbo Dong
- Department of Chemical Engineering, West Virginia University, Morgantown, WV 26505, USA
| | - Bing Shen
- Department of Physiology, Anhui Medical University, Hefei, Anhui 230032, P. R. China
| |
Collapse
|
87
|
Liposomes as carriers of hydrophilic small molecule drugs: Strategies to enhance encapsulation and delivery. Colloids Surf B Biointerfaces 2014; 123:345-63. [DOI: 10.1016/j.colsurfb.2014.09.029] [Citation(s) in RCA: 292] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 07/30/2014] [Accepted: 09/14/2014] [Indexed: 12/18/2022]
|
88
|
Ryan GM, Kaminskas LM, Porter CJ. Nano-chemotherapeutics: Maximising lymphatic drug exposure to improve the treatment of lymph-metastatic cancers. J Control Release 2014; 193:241-56. [DOI: 10.1016/j.jconrel.2014.04.051] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/21/2014] [Accepted: 04/23/2014] [Indexed: 01/17/2023]
|
89
|
Abstract
In recent years, hundreds of genes have been linked to a variety of human diseases, and the field of gene therapy has emerged as a way to treat this wide range of diseases. The main goal of gene therapy is to find a gene delivery vehicle that can successfully target diseased cells and deliver therapeutic genes directly to their cellular compartment. The two main types of gene delivery vectors currently being investigated in clinical trials are recombinant viral vectors and synthetic nonviral vectors. Recombinant viral vectors take advantage of the evolutionarily optimized viral mechanisms to deliver genes, but they can be hard to specifically target in vivo and are also associated with serious side effects. Synthetic nonviral vectors are made out of highly biocompatible lipids or polymers, but they are much less efficient at delivering their genetic payload due to the lack of any active delivery mechanism. This mini review will introduce the current state of gene delivery in clinical trials, and discuss the specific challenges associated with each of these vectors. It will also highlight some specific gaps in knowledge that are limiting the advancement of this field and touch on the current areas of research being explored to overcome them.
Collapse
Affiliation(s)
- Yarong Liu
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA
| | - Jennifer Rohrs
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
90
|
Jin S, Li S, Wang C, Liu J, Yang X, Wang PC, Zhang X, Liang XJ. Biosafe nanoscale pharmaceutical adjuvant materials. J Biomed Nanotechnol 2014; 10:2393-419. [PMID: 25429253 PMCID: PMC4242152 DOI: 10.1166/jbn.2014.1898] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Thanks to developments in the field of nanotechnology over the past decades, more and more biosafe nanoscale materials have become available for use as pharmaceutical adjuvants in medical research. Nanomaterials possess unique properties which could be employed to develop drug carriers with longer circulation time, higher loading capacity, better stability in physiological conditions, controlled drug release, and targeted drug delivery. In this review article, we will review recent progress in the application of representative organic, inorganic and hybrid biosafe nanoscale materials in pharmaceutical research, especially focusing on nanomaterial-based novel drug delivery systems. In addition, we briefly discuss the advantages and notable functions that make these nanomaterials suitable for the design of new medicines; the biosafety of each material discussed in this article is also highlighted to provide a comprehensive understanding of their adjuvant attributes.
Collapse
Affiliation(s)
- Shubin Jin
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Shengliang Li
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Department of Neurobiology and Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, P. R. China
| | - Chongxi Wang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Juan Liu
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Xiaolong Yang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Paul C. Wang
- Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington DC 20060, USA
| | - Xin Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| |
Collapse
|
91
|
Tiantian Y, Wenji Z, Mingshuang S, Rui Y, Shuangshuang S, Yuling M, Jianhua Y, Xinggang Y, Shujun W, Weisan P. Study on intralymphatic-targeted hyaluronic acid-modified nanoliposome: Influence of formulation factors on the lymphatic targeting. Int J Pharm 2014; 471:245-57. [DOI: 10.1016/j.ijpharm.2014.05.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 04/27/2014] [Accepted: 05/18/2014] [Indexed: 10/25/2022]
|
92
|
Advances in imaging probes and optical microendoscopic imaging techniques for early in vivo cancer assessment. Adv Drug Deliv Rev 2014; 74:53-74. [PMID: 24120351 DOI: 10.1016/j.addr.2013.09.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 09/18/2013] [Accepted: 09/27/2013] [Indexed: 12/12/2022]
Abstract
A new chapter in the history of medical diagnosis happened when the first X-ray technology was invented in the late 1800s. Since then, many non-invasive and minimally invasive imaging techniques have been invented for clinical diagnosis to research in cellular biology, drug discovery, and disease monitoring. These imaging modalities have leveraged the benefits of significant advances in computer, electronics, and information technology and, more recently, targeted molecular imaging. The development of targeted contrast agents such as fluorescent and nanoparticle probes coupled with optical imaging techniques has made it possible to selectively view specific biological events and processes in both in vivo and ex vivo systems with great sensitivity and selectivity. Thus, the combination of targeted molecular imaging probes and optical imaging techniques have become a mainstay in modern medicinal and biological research. Many promising results have demonstrated great potentials to translate to clinical applications. In this review, we describe a discussion of employing imaging probes and optical microendoscopic imaging techniques for cancer diagnosis.
Collapse
|
93
|
Chen C, Zhou JL, Han X, Song F, Wang XL, Wang YZ. A prodrug strategy based on chitosan for efficient intracellular anticancer drug delivery. NANOTECHNOLOGY 2014; 25:255101. [PMID: 24896540 DOI: 10.1088/0957-4484/25/25/255101] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Doxorubicin (DOX), one of the most widely used anticancer drugs, is restricted in clinical application due to its severe side effects and inefficient cellular uptake. To overcome the drawbacks, herein, an endosomal pH-activated prodrug was designed and fabricated by conjugating DOX with chitosan via an acid-cleavable hydrazone bond. The resulting DOX conjugates can self-assemble into nano-sized particles, which were very stable and presented no burst release of DOX at a neutral pH condition. Notably, the nanoparticles exhibited excellent cell uptake properties and a remarkable drug accumulation in tumor cells. Once internalized into the cells, moreover, DOX can be fast released from the nanoparticles, and the release mechanism changed from the anomalous transport at pH 7.4 to the combination pattern of diffusion- and erosion-controlled release at pH 6.0 or 5.0. The prodrugs showed obvious cytotoxicity for HeLa cells with fairly low IC50 values, offering a new platform for targeted cancer therapy.
Collapse
Affiliation(s)
- Cheng Chen
- Center for Degradable and Flame-Retardant Polymeric Materials (ERCPM-MoE), College of Chemistry, State Key Laboratory of Polymer Materials Engineering, National Engineering Laboratory of Eco-Friendly Polymeric Materials (Sichuan), Sichuan University, 29 Wangjiang Road, Chengdu 610064, People's Republic of China
| | | | | | | | | | | |
Collapse
|
94
|
Wei X, Zhan C, Chen X, Hou J, Xie C, Lu W. Retro-inverso isomer of Angiopep-2: a stable d-peptide ligand inspires brain-targeted drug delivery. Mol Pharm 2014; 11:3261-8. [PMID: 24673510 DOI: 10.1021/mp500086e] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The blood-brain barrier (BBB) prevents most drugs from reaching the site of central nervous system (CNS) diseases, intensively confining the therapeutic efficiency. Angiopep-2 (here termed (L)Angiopep), which is a 19-mer peptide derived from human Kunitz domain, can trigger transcytosis and traverse the BBB by recognizing low density lipoprotein-related protein 1 (LRP-1) expressed on the brain capillary endothelial cells. Various enzymes in the blood and the BBB, however, present multiple metabolic barriers to peptide-inspired brain-targeted drug delivery. Here we designed a retro-inverso isomer of (L)Angiopep, termed (D)Angiopep, to inspire brain-targeted drug delivery. Both (D)Angiopep and (L)Angiopep displayed high uptake capacity in LRP-1 overexpressed cells, including bEnd.3 and U87 cells. (D)Angiopep demonstrated lower uptake efficiency in both cell lines than did (L)Angiopep, suggestive of lower binding affinity to LRP-1 of the d-peptide. (D)Angiopep was resistant to proteolysis in fresh rat blood serum, while more than 85% of (L)Angiopep disappeared within 2 h. Endocytosed (D)Angiopep and (L)Angiopep were found to be colocalized with lysosomal compartments of bEnd.3 cells, indicating that susceptibility to proteolysis of (L)Angiopep in the BBB may further attenuate its transcytosis efficiency. In vivo, (D)Angiopep modified PEG-DSPE micelles displayed high distribution in normal brain and intracranial glioblastoma. Due to the expression of LRP-1 on the BBB and glioblastoma cells, proteolytically stable (D)Angiopep holds much potential for designing two-order brain tumor targeted delivery systems.
Collapse
Affiliation(s)
- Xiaoli Wei
- Department of Pharmaceutics, School of Pharmacy, Fudan University , Shanghai, 201203, P. R. China
| | | | | | | | | | | |
Collapse
|
95
|
Yang C, Fu ZX. Liposomal delivery and polyethylene glycol-liposomal oxaliplatin for the treatment of colorectal cancer (Review). Biomed Rep 2014; 2:335-339. [PMID: 24748970 PMCID: PMC3990200 DOI: 10.3892/br.2014.249] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 02/25/2014] [Indexed: 12/17/2022] Open
Abstract
Oxaliplatin is effective for the treatment of advanced colorectal cancer; however, its application is restricted due to its dose-limiting toxicity. Liposomes are sphere-shaped vesicles consisting of one or more phospholipid bilayers. Liposomes as drug carriers are characterized by delayed release, lesion targeting and may be used as a drug-delivery system to decrease the side effects of cytotoxic drugs. Active targeting modification of liposomes may change the biological distribution of the anticancer agents, reduce or reverse multidrug resistance of tumor cells and enhance the effects of anticancer therapy. Based on the characteristics mentioned above, the aim of the present review was to demonstrate that polyethylene glycol-liposomes containing oxaliplatin may offer advantages for the treatment of colorectal cancer in clinical practice.
Collapse
Affiliation(s)
- Chuang Yang
- Department of General Surgery, Third People's Hospital of Mianyang, Mianyang, Sichuan 621000, P.R. China ; Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing, Chongqing 400016, P.R. China
| | - Zhong-Xue Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing, Chongqing 400016, P.R. China
| |
Collapse
|
96
|
Singh I, Swami R, Khan W, Sistla R. Lymphatic system: a prospective area for advanced targeting of particulate drug carriers. Expert Opin Drug Deliv 2013; 11:211-29. [PMID: 24350774 DOI: 10.1517/17425247.2014.866088] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The lymphatic system has a critical role in the immune system's recognition and response to disease and it is an additional circulatory system throughout the entire body. Extensive multidisciplinary investigations have been carried out in the area of lymphatic delivery, and lymphatic targeting has attracted a lot of attention for providing preferential chemotherapy and improving bioavailability of drugs that undergo hepatic first-pass metabolism. AREAS COVERED This review focuses on progress in the field of lymphatic therapeutics and diagnosis. Moreover, the anatomy and physiology of the lymphatic system, particulate drug carriers and different physicochemical parameters of both modified and unmodified particulate drug carriers and their effect on lymphatic targeting are addressed. EXPERT OPINION Particulate drug carriers have encouraged lymphatic targeting, but there are still challenges in targeting drugs and bioactives to specific sites, maintaining desired action and crossing all the physiological barriers. Lymphatic therapy using drug-encapsulated lipid carriers, especially liposomes and solid lipid nanoparticles, emerges as a new technology to provide better penetration into the lymphatics where residual disease exists. Size is the most important criteria when designing nanocarriers for targeting lymphatic vessels as the transportation of these particles into lymphatic vessels is size dependent. By increasing our understanding of lymphatic transport and uptake, and the role of lymphatics in various diseases, we can design new therapeutics for effective disease control.
Collapse
Affiliation(s)
- Indu Singh
- National Institute of Pharmaceutical Education & Research (NIPER), Department of Pharmaceutics , Hyderabad 500037 , India +91 40 27193004, +91 40 23073741 ; +91 40 27193753, +91 40 23073751 ; ;
| | | | | | | |
Collapse
|
97
|
Yan Z, Yang Y, Wei X, Zhong J, Wei D, Liu L, Xie C, Wang F, Zhang L, Lu W, He D. Tumor-Penetrating Peptide Mediation: An Effective Strategy for Improving the Transport of Liposomes in Tumor Tissue. Mol Pharm 2013; 11:218-25. [DOI: 10.1021/mp400393a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Zhiqiang Yan
- National Engineering Research Center for Nanotechnology, Shanghai 200241, P.R. China
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics
and New Drug Development, Institutes for
Advanced Interdisciplinary Research, East China Normal University, Shanghai 200062, P.R. China
| | - Yiyi Yang
- National Engineering Research Center for Nanotechnology, Shanghai 200241, P.R. China
- School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Xiaoli Wei
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department
of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, P.R. China
| | - Jian Zhong
- National Engineering Research Center for Nanotechnology, Shanghai 200241, P.R. China
| | - Daixu Wei
- National Engineering Research Center for Nanotechnology, Shanghai 200241, P.R. China
| | - Lu Liu
- National Engineering Research Center for Nanotechnology, Shanghai 200241, P.R. China
| | - Cao Xie
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department
of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, P.R. China
| | - Fei Wang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department
of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, P.R. China
| | - Lin Zhang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department
of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, P.R. China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department
of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, P.R. China
| | - Dannong He
- National Engineering Research Center for Nanotechnology, Shanghai 200241, P.R. China
- School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| |
Collapse
|
98
|
Johansson A, Hamzah J, Ganss R. License for destruction: tumor-specific cytokine targeting. Trends Mol Med 2013; 20:16-24. [PMID: 24169116 DOI: 10.1016/j.molmed.2013.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 09/27/2013] [Accepted: 10/02/2013] [Indexed: 12/28/2022]
Abstract
Stroma is an integral part of solid tumors and plays a key role in growth promotion and immune suppression. Most current therapies focus on destroying tumors and/or abnormal vasculature. However, evidence is emerging that anticancer efficacy improves with vessel normalization rather than destruction. Specific targeting of cytokines into tumors provides proof-of-concept that tumor stroma is dynamic and can be remodeled to increase drug access and alleviate immune suppression. Changing the inflammatory milieu 'opens' tumors for therapy and thus provides a license for destruction. This involves reprogramming of paracrine signaling networks between multiple stromal components to break the vicious cycle of angiogenesis and immune suppression. With active immunotherapy rapidly moving into the clinic, local cytokine delivery emerges as an attractive adjuvant.
Collapse
Affiliation(s)
- Anna Johansson
- Western Australian Institute for Medical Research, University of Western Australia, Centre for Medical Research, Perth, 6000, Australia
| | - Juliana Hamzah
- Western Australian Institute for Medical Research, University of Western Australia, Centre for Medical Research, Perth, 6000, Australia
| | - Ruth Ganss
- Western Australian Institute for Medical Research, University of Western Australia, Centre for Medical Research, Perth, 6000, Australia.
| |
Collapse
|
99
|
A noticeable phenomenon: thiol terminal PEG enhances the immunogenicity of PEGylated emulsions injected intravenously or subcutaneously into rats. Eur J Pharm Biopharm 2013; 85:744-51. [PMID: 24129310 DOI: 10.1016/j.ejpb.2013.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 09/08/2013] [Accepted: 10/04/2013] [Indexed: 12/20/2022]
Abstract
Repeated intravenous injection of long-circulating methoxy-polyethylene glycol (PEG)-liposomes alters the pharmacokinetics and biodistribution of the second administration, regarded as the "accelerated blood clearance (ABC) phenomenon." Nevertheless, the effect of terminal groups of distearoylphosphatidylethanolamine-polyethylene glycol (DSPE-PEG) on the induction of the ABC phenomenon had not been reported previously. In this study, rats were injected intravenously or subcutaneously with PEG coated emulsions (DE) which were prepared using PEG terminated with either the methoxyl (OCH3), hydroxyl (OH), amino (NH2), carboxyl (COOH), or thiol (SH) group. DE-OCH3 demonstrated the longest prolonged half-life in vivo after a single intravenous injection, followed by DE-SH and DE-COOH. In contrast, DE-OH was rapidly removed from the blood circulation, as was DE-NH2. Moreover, we observed a strong positive relationship between the circulation time of initially injected PEGylated emulsions and the extent to which the ABC phenomenon was induced, but a exception of DE-SH increasing the ABC effect. Furthermore, the present study suggested that thiols might stimulate the proliferation and differentiation of B cells to induce the fastest clearance of the second intravenous administration by inducing the synthesis of the cell membrane and cytosolic proteins or reacting with follicular dendritic cells. The results strongly suggested that thiol groups played a stimulatory role in the immune response and provided a considerable implication for multiple drug therapy of thiol groups.
Collapse
|
100
|
Yang Y, Yan Z, Wei D, Zhong J, Liu L, Zhang L, Wang F, Wei X, Xie C, Lu W, He D. Tumor-penetrating peptide functionalization enhances the anti-glioblastoma effect of doxorubicin liposomes. NANOTECHNOLOGY 2013; 24:405101. [PMID: 24029287 DOI: 10.1088/0957-4484/24/40/405101] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The targeted therapeutic effect of nano drug delivery system for glioblastoma has been hampered by the weak enhanced permeability and retention (EPR) effect of glioblastoma and the low delivering efficiency of NDDS in glioblastoma tissue. In this study, a tumor-penetrating peptide (RGERPPR), the specific ligand of neuropilin-1 overexpressed on glioblastoma and endothelial cells, was used as a targeting moiety to enhance the anti-glioblastoma effect of doxorubicin liposomes. Firstly, RGERPPR-PEG-DSPE was synthesized and used to prepare the RGERPPR peptide-functionalized liposomes (RGE-LS), which showed vesicle sizes of around 90 nm and narrow size distributions. The cellular uptake and in vivo near-infrared fluorescence imaging test displayed that RGE-LS exhibited increased uptake by glioblastoma cells and intracranial glioblastoma tissues. The cytotoxicity assay and anti-glioblastoma study proved that RGERPPR functionalization significantly enhanced the in vitro inhibitory effect of doxorubicin liposomes on glioblastoma cells and prolonged the median survival time of nude mice bearing intracranial glioblastoma. Finally, the immunofluorescence analysis evidenced that RGE-LS were able to penetrate through tumor vessels and stroma and deep into the whole tumor tissue. The results indicated that tumor-penetrating peptide functionalization is an effective strategy for enhancing the anti-glioblastoma effect of doxorubicin liposomes.
Collapse
Affiliation(s)
- Yiyi Yang
- School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China. National Engineering Research Center for Nanotechnology, Shanghai 200241, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|