51
|
Xu G, Lou N, Xu Y, Shi H, Ruan H, Xiao W, Liu L, Xiao H, Qiu B, Bao L, Yuan C, Chen K, Yang H, Zhang X. Diagnostic and prognostic value of scavenger receptor class B type 1 in clear cell renal cell carcinoma. Tumour Biol 2017; 39:1010428317699110. [PMID: 28466781 DOI: 10.1177/1010428317699110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Aberrant expression of scavenger receptor class B type 1 has been reported in several human cancers. Nevertheless, the roles of scavenger receptor class B type 1 in clear cell renal cell carcinoma remain unclear. The aim of this study was to evaluate the diagnostic and prognostic value of scavenger receptor class B type 1 in clear cell renal cell carcinoma. The messenger RNA level of scavenger receptor class B type 1 in clear cell renal cell carcinoma tissues was detected by quantitative reverse transcription polymerase chain reaction, while protein level was determined by western blot and immunohistochemistry. The lipid content between clear cell renal cell carcinoma tissues and normal kidney tissues was differentiated by Oil Red O and hematoxylin-eosin staining. The diagnostic value of scavenger receptor class B type 1 was determined by receiver operating characteristic curve. The prognostic significance of scavenger receptor class B type 1 was assessed by Kaplan-Meier analysis and Cox regression analysis. Our results showed that the expression of scavenger receptor class B type 1 in clear cell renal cell carcinoma tissues at both messenger RNA and protein level was much higher than that in normal kidney tissues. Receiver operating characteristic curve analysis exhibited a significant value of area under the curve (0.8486, 95% confidence interval: 0.7926-0.9045) with strong sensitivity (0.75, 95% confidence interval: 0.6535-0.8312) and specificity (0.90, 95% confidence interval: 0.8238-0.9510). Kaplan-Meier analysis revealed that patients with higher scavenger receptor class B type 1 expression had shorter progression-free survival time. Cox analysis indicated that scavenger receptor class B type 1 was an independent prognostic biomarker. In conclusion, our findings implied that scavenger receptor class B type 1 might serve as a diagnostic and independent prognostic biomarker in clear cell renal cell carcinoma.
Collapse
Affiliation(s)
- Guanghua Xu
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Lou
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuchen Xu
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hangchuan Shi
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hailong Ruan
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Xiao
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Liu
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haibing Xiao
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Qiu
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Bao
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changfei Yuan
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Chen
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Yang
- 2 Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Zhang
- 1 Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
52
|
Effect of size and pegylation of liposomes and peptide-based synthetic lipoproteins on tumor targeting. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1869-1878. [PMID: 28434931 DOI: 10.1016/j.nano.2017.04.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 03/28/2017] [Accepted: 04/10/2017] [Indexed: 12/31/2022]
Abstract
Synthetic high-density lipoprotein nanoparticles (sHDL) are a valuable class of nanomedicines with established animal safety profile, clinical tolerability and therapeutic efficacy for cardiovascular applications. In this study we examined how the scavenger receptor B-I-mediated (SR-BI) tumor-targeting ability of sHDL, long plasma circulation half-life, and small particle size (9.6±0.2nm) impacted sHDL accumulation in SR-BI positive colorectal carcinoma cells, 3D tumor spheroids, and in vivo xenografts. We compared tumor accumulation of sHDL with that of liposomes (LIP, 130.7±0.8nm), pegylated liposomes (PEG-LIP, 101±2nm), and pegylated sHDL (12.1±0.1nm), all prepared with the same lipid components. sHDL penetrated deep (210μm) into tumor spheroids and exhibited 12- and 3-fold higher in vivo solid tumor accumulation, compared with LIP (p<0.01) and PEG-LIP (p<0.05), respectively. These results suggest that sHDL with established human safety possess promising intrinsic tumor-targeted properties.
Collapse
|
53
|
Mooberry LK, Sabnis NA, Panchoo M, Nagarajan B, Lacko AG. Targeting the SR-B1 Receptor as a Gateway for Cancer Therapy and Imaging. Front Pharmacol 2016; 7:466. [PMID: 28018216 PMCID: PMC5156841 DOI: 10.3389/fphar.2016.00466] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/16/2016] [Indexed: 01/25/2023] Open
Abstract
Malignant tumors display remarkable heterogeneity to the extent that even at the same tissue site different types of cells with varying genetic background may be found. In contrast, a relatively consistent marker the scavenger receptor type B1 (SR-B1) has been found to be consistently overexpressed by most tumor cells. Scavenger Receptor Class B Type I (SR-BI) is a high density lipoprotein (HDL) receptor that facilitates the uptake of cholesterol esters from circulating lipoproteins. Additional findings suggest a critical role for SR-BI in cholesterol metabolism, signaling, motility, and proliferation of cancer cells and thus a potential major impact in carcinogenesis and metastasis. Recent findings indicate that the level of SR-BI expression correlate with aggressiveness and poor survival in breast and prostate cancer. Moreover, genomic data show that depending on the type of cancer, high or low SR-BI expression may promote poor survival. This review discusses the importance of SR-BI as a diagnostic as well as prognostic indicator of cancer to help elucidate the contributions of this protein to cancer development, progression, and survival. In addition, the SR-B1 receptor has been shown to serve as a potential gateway for the delivery of therapeutic agents when reconstituted high density lipoprotein nanoparticles are used for their transport to cancer cells and tumors. Opportunities for the development of new technologies, particularly in the areas of cancer therapy and tumor imaging are discussed.
Collapse
Affiliation(s)
- Linda K. Mooberry
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort WorthTX, USA
| | - Nirupama A. Sabnis
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort WorthTX, USA
| | - Marlyn Panchoo
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort WorthTX, USA
| | - Bhavani Nagarajan
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort WorthTX, USA
| | - Andras G. Lacko
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort WorthTX, USA
- Department of Pediatrics, University of North Texas Health Science Center, Fort WorthTX, USA
| |
Collapse
|
54
|
Yuan Y, Wen J, Tang J, Kan Q, Ackermann R, Olsen K, Schwendeman A. Synthetic high-density lipoproteins for delivery of 10-hydroxycamptothecin. Int J Nanomedicine 2016; 11:6229-6238. [PMID: 27920529 PMCID: PMC5125756 DOI: 10.2147/ijn.s112835] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The purpose of this study was to develop a novel synthetic high-density lipoprotein (sHDL) nanoparticle delivery system for 10-hydroxycamptothecin (HCPT) for treatment of colon carcinoma. HDL is recognized by scavenger receptor B-I (SR-BI) over-expressed in colon carcinomas 5- to 35-fold relative to the human fibroblasts. The sHDL nanoparticles were composed of apolipoprotein A-I mimic peptide (5A) and contained 0.5%–1.5% (w/w) of HCPT. An optimized HCPT-sHDL formulation exhibited 0.7% HCPT loading with 70% efficiency with an average size of 10–12 nm. Partitioning of HCPT in the sHDL lipid membrane enhanced drug stability in its active lactone form, increased solubilization, and enabled slow release. Cytotoxicity studies in HT29 colon carcinoma cells revealed that the IC50 of HCPT-sHDL was approximately 3-fold lower than that of free HCPT. Pharmacokinetics in rats following intravenous administration showed that the area under the serum concentration-time curve (AUC0−t) and Cmax of HCPT-HDL were 2.7- and 6.5-fold higher relative to the values for the free HCPT, respectively. These results suggest that sHDL-based formulations of hydrophobic drugs are useful for future evaluation in treatment of SR-BI-positive tumors.
Collapse
Affiliation(s)
- Yue Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China; Department of Pharmaceutical Sciences, Biointerfaces Institute, College of Pharmacy, University of Michigan
| | - Jian Wen
- Department of Internal Medicine, Division of Molecular Medicine and Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jie Tang
- Department of Pharmaceutical Sciences, Biointerfaces Institute, College of Pharmacy, University of Michigan
| | - Qiming Kan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Rose Ackermann
- Department of Pharmaceutical Sciences, Biointerfaces Institute, College of Pharmacy, University of Michigan
| | - Karl Olsen
- Department of Pharmaceutical Sciences, Biointerfaces Institute, College of Pharmacy, University of Michigan
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, Biointerfaces Institute, College of Pharmacy, University of Michigan
| |
Collapse
|
55
|
Mo ZC, Ren K, Liu X, Tang ZL, Yi GH. A high-density lipoprotein-mediated drug delivery system. Adv Drug Deliv Rev 2016; 106:132-147. [PMID: 27208399 DOI: 10.1016/j.addr.2016.04.030] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/13/2016] [Accepted: 04/27/2016] [Indexed: 01/08/2023]
Abstract
High-density lipoprotein (HDL) is a comparatively dense and small lipoprotein that can carry lipids as a multifunctional aggregate in plasma. Several studies have shown that increasing the levels or improving the functionality of HDL is a promising target for treating a wide variety of diseases. Among lipoproteins, HDL particles possess unique physicochemical properties, including naturally synthesized physiological components, amphipathic apolipoproteins, lipid-loading and hydrophobic agent-incorporating characteristics, specific protein-protein interactions, heterogeneity, nanoparticles, and smaller size. Recently, the feasibility and superiority of using HDL particles as drug delivery vehicles have been of great interest. In this review, we summarize the structure, constituents, biogenesis, remodeling, and reconstitution of HDL drug delivery systems, focusing on their delivery capability, characteristics, applications, manufacturing, and drug-loading and drug-targeting characteristics. Finally, the future prospects are presented regarding the clinical application and challenges of using HDL as a pharmacodelivery carrier.
Collapse
Affiliation(s)
- Zhong-Cheng Mo
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang City 421001, Hunan Province, China; Department of Histology and Embryology, University of South China, Hengyang, Hunan 421001, China
| | - Kun Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang City 421001, Hunan Province, China
| | - Xing Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 100005 Beijing, China
| | - Zhen-Li Tang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang City 421001, Hunan Province, China
| | - Guang-Hui Yi
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang City 421001, Hunan Province, China.
| |
Collapse
|
56
|
Yu R, Lv Y, Wang J, Pan N, Zhang R, Wang X, Yu H, Tan L, Zhao Y, Li B. Baicalin promotes cholesterol efflux by regulating the expression of SR-BI in macrophages. Exp Ther Med 2016; 12:4113-4120. [PMID: 28105139 DOI: 10.3892/etm.2016.3884] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 10/05/2016] [Indexed: 01/22/2023] Open
Abstract
Intake of a high dosage of baicalin has previously been shown to attenuate hyperlipidemia induced by a high-fat diet. Baicalin functions as an activator of peroxisome proliferator-activated receptor-γ (PPAR-γ), which is the key regulator of reverse cholesterol transport (RCT). The present study aimed to test the hypothesis that baicalin could promote cholesterol efflux in macrophages through activating PPAR-γ. Phorbol 12-myristate 13-acetate-stimulated THP-1 cells were treated with oxidized low-density lipoprotein and (3H)-cholesterol for 24 h, and the effects of baicalin on cholesterol efflux were evaluated in the presence of apolipoprotein A-1 (ApoA-1), or high-density lipoprotein subfraction 2 (HDL2) or subfraction 3 (HDL3). The expression levels of scavenger receptor class B type I (SR-BI), PPAR-γ and liver X receptor-α (LXRα) were detected and specific inhibitors or activators of SR-BI, PPAR-γ and LXRα were applied to investigate the mechanism. Treatment of THP-1 macrophages with baicalin significantly accelerated HDL-mediated, but not ApoA-1-mediated cholesterol efflux. However, baicalin treatment increased the expression of SR-BI at the mRNA and protein levels in a dose- and time-dependent manner, and pre-treatment with the SR-BI inhibitor BLT-1 and SR-BI small interfering RNA significantly inhibited baicalin-induced cholesterol efflux. Furthermore, baicalin increased the expression of PPAR-γ and LXRα, and the application of specific agonists and inhibitors of PPAR-γ and LXRα changed the expression of SR-BI, as well as cholesterol efflux. It may be concluded that baicalin induced cholesterol efflux from THP-1 macrophages via the PPAR-γ/LXRα/SR-BI pathway.
Collapse
Affiliation(s)
- Renchao Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Yuexia Lv
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Juanling Wang
- Clinical Skill Training Center, People's Hospital of Weifang, Weifang, Shandong 261041, P.R. China
| | - Nana Pan
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Rui Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Xiaxia Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Lijuan Tan
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Yunhe Zhao
- Department of Cardiology, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| | - Bo Li
- Department of Cardiology, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
57
|
Gutierrez-Pajares JL, Ben Hassen C, Chevalier S, Frank PG. SR-BI: Linking Cholesterol and Lipoprotein Metabolism with Breast and Prostate Cancer. Front Pharmacol 2016; 7:338. [PMID: 27774064 PMCID: PMC5054001 DOI: 10.3389/fphar.2016.00338] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022] Open
Abstract
Studies have demonstrated the significant role of cholesterol and lipoprotein metabolism in the progression of cancer. The SCARB1 gene encodes the scavenger receptor class B type I (SR-BI), which is an 82-kDa glycoprotein with two transmembrane domains separated by a large extracellular loop. SR-BI plays an important role in the regulation of cholesterol exchange between cells and high-density lipoproteins. Accordingly, hepatic SR-BI has been shown to play an essential role in the regulation of the reverse cholesterol transport pathway, which promotes the removal and excretion of excess body cholesterol. In the context of atherosclerosis, SR-BI has been implicated in the regulation of intracellular signaling, lipid accumulation, foam cell formation, and cellular apoptosis. Furthermore, since lipid metabolism is a relevant target for cancer treatment, recent studies have focused on examining the role of SR-BI in this pathology. While signaling pathways have initially been explored in non-tumoral cells, studies with cancer cells have now demonstrated SR-BI's function in tumor progression. In this review, we will discuss the role of SR-BI during tumor development and malignant progression. In addition, we will provide insights into the transcriptional and post-transcriptional regulation of the SCARB1 gene. Overall, studying the role of SR-BI in tumor development and progression should allow us to gain useful information for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Jorge L Gutierrez-Pajares
- Université François Rabelais de Tours, Faculté de Médecine-INSERM UMR1069 "Nutrition, Croissance et Cancer" Tours, France
| | - Céline Ben Hassen
- Université François Rabelais de Tours, Faculté de Médecine-INSERM UMR1069 "Nutrition, Croissance et Cancer" Tours, France
| | - Stéphan Chevalier
- Université François Rabelais de Tours, Faculté de Médecine-INSERM UMR1069 "Nutrition, Croissance et Cancer" Tours, France
| | - Philippe G Frank
- Université François Rabelais de Tours, Faculté de Médecine-INSERM UMR1069 "Nutrition, Croissance et Cancer" Tours, France
| |
Collapse
|
58
|
Simonsen JB. Evaluation of reconstituted high-density lipoprotein (rHDL) as a drug delivery platform – a detailed survey of rHDL particles ranging from biophysical properties to clinical implications. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2161-2179. [DOI: 10.1016/j.nano.2016.05.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 12/15/2022]
|
59
|
Rajora MA, Zheng G. Targeting SR-BI for Cancer Diagnostics, Imaging and Therapy. Front Pharmacol 2016; 7:326. [PMID: 27729859 PMCID: PMC5037127 DOI: 10.3389/fphar.2016.00326] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/06/2016] [Indexed: 01/13/2023] Open
Abstract
Scavenger receptor class B type I (SR-BI) plays an important role in trafficking cholesteryl esters between the core of high density lipoprotein and the liver. Interestingly, this integral membrane protein receptor is also implicated in the metabolism of cholesterol by cancer cells, whereby overexpression of SR-BI has been observed in a number of tumors and cancer cell lines, including breast and prostate cancers. Consequently, SR-BI has recently gained attention as a cancer biomarker and exciting target for the direct cytosolic delivery of therapeutic agents. This brief review highlights these key developments in SR-BI-targeted cancer therapies and imaging probes. Special attention is given to the exploration of high density lipoprotein nanomimetic platforms that take advantage of upregulated SR-BI expression to facilitate targeted drug-delivery and cancer diagnostics, and promising future directions in the development of these agents.
Collapse
Affiliation(s)
- Maneesha A Rajora
- Princess Margaret Cancer Centre and Techna Institute, University Health NetworkToronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of TorontoToronto, ON, Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre and Techna Institute, University Health NetworkToronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of TorontoToronto, ON, Canada; Department of Medical Biophysics, University of TorontoToronto, ON, Canada
| |
Collapse
|
60
|
Jin H, Qian Y, Dai Y, Qiao S, Huang C, Lu L, Luo Q, Chen J, Zhang Z. Magnetic Enrichment of Dendritic Cell Vaccine in Lymph Node with Fluorescent-Magnetic Nanoparticles Enhanced Cancer Immunotherapy. Am J Cancer Res 2016; 6:2000-2014. [PMID: 27698936 PMCID: PMC5039339 DOI: 10.7150/thno.15102] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/13/2016] [Indexed: 01/08/2023] Open
Abstract
Dendritic cell (DC) migration to the lymph node is a key component of DC-based immunotherapy. However, the DC homing rate to the lymphoid tissues is poor, thus hindering the DC-mediated activation of antigen-specific T cells. Here, we developed a system using fluorescent magnetic nanoparticles (α-AP-fmNPs; loaded with antigen peptide, iron oxide nanoparticles, and indocyanine green) in combination with magnetic pull force (MPF) to successfully manipulate DC migration in vitro and in vivo. α-AP-fmNPs endowed DCs with MPF-responsiveness, antigen presentation, and simultaneous optical and magnetic resonance imaging detectability. We showed for the first time that α-AP-fmNP-loaded DCs were sensitive to MPF, and their migration efficiency could be dramatically improved both in vitro and in vivo through MPF treatment. Due to the enhanced migration of DCs, MPF treatment significantly augmented antitumor efficacy of the nanoparticle-loaded DCs. Therefore, we have developed a biocompatible approach with which to improve the homing efficiency of DCs and subsequent anti-tumor efficacy, and track their migration by multi-modality imaging, with great potential applications for DC-based cancer immunotherapy.
Collapse
|
61
|
Qian Y, Jin H, Qiao S, Dai Y, Huang C, Lu L, Luo Q, Zhang Z. Targeting dendritic cells in lymph node with an antigen peptide-based nanovaccine for cancer immunotherapy. Biomaterials 2016; 98:171-83. [PMID: 27192420 DOI: 10.1016/j.biomaterials.2016.05.008] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/16/2016] [Accepted: 05/03/2016] [Indexed: 12/21/2022]
Abstract
The design of peptide-based subunit vaccine formulations for the direct delivery of tumor antigen peptides (Aps) to dendritic cells (DCs) localized within draining lymph nodes (DLNs) is challenging. Mature DCs (mDCs) are abundantly distributed within DLNs but have dramatically reduced endocytic uptake and antigen-processing abilities, so their role as potential vaccine targets has been largely overlooked. Here we report an ultra-small biocompatible nanovaccine (α-Ap-FNP) functionalized by avidly targeting delivery of Ap via the scavenger receptor class B1 (SR-B1) pathway to mDCs. The self-assembly, small size (∼30 nm), SR-B1-targeting and optical properties of α-Ap-FNP resulted in its efficient Ap loading, substantial LN accumulation, targeting of mDCs and enhanced Ap presentation, and fluorescence trafficking, respectively. We also demonstrate that the α-Ap-FNP can be either used alone or encapsulated with CpG oligodeoxynucleotide as a prophylactic and therapeutic vaccine. Thus, the excellent properties of α-Ap-FNP provide it potential for clinical applications as a potent nanovaccine for cancer immunotherapy.
Collapse
Affiliation(s)
- Yuan Qian
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Honglin Jin
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Sha Qiao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yanfeng Dai
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chuan Huang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lisen Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
62
|
Challenges and opportunities for siRNA-based cancer treatment. Cancer Lett 2016; 387:77-83. [PMID: 27045474 DOI: 10.1016/j.canlet.2016.03.045] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 12/14/2022]
Abstract
As one of the life-threatening diseases involving multi-step genetic and epigenetic disorders, cancer has long been a dynamic research area for siRNA-based therapy as half of the current siRNA-based clinical trials are involved in oncology. However, despite consistent enthusiasm in the academic world, siRNA-based cancer treatment still faces obstacles and difficulties in clinical development. In this article, we discuss key challenges facing siRNA-based cancer treatment revealed from recent clinical and preclinical studies, including chemical modification, tumour penetration, endosomal escape, target selection and off-target effects. In addition, opportunities and avenues for translating siRNA technology from bench to oncologic clinics are explored.
Collapse
|
63
|
Synthetic high-density lipoprotein nanoparticles: A novel therapeutic strategy for adrenocortical carcinomas. Surgery 2015; 159:284-94. [PMID: 26582501 DOI: 10.1016/j.surg.2015.08.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 07/20/2015] [Accepted: 08/12/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chemotherapeutic strategies for adrenocortical carcinoma (ACC) carry substantial toxicities. Cholesterol is critical for ACC cell growth and steroidogenesis, and ACC cells overexpress scavenger receptor BI, which uptakes cholesterol from circulating high-density lipoprotein (HDL) cholesterol. We hypothesize that cholesterol-free synthetic-HDL nanoparticles (sHDL) will deplete cholesterol and synergize with chemotherapeutics to achieve enhanced anticancer effects at lesser (less toxic) drug levels. METHODS The antiproliferative efficacy of ACC cells for the combinations of sHDL with chemotherapeutics was tested by Cell-Titer Glo. Cortisol levels were measured from the culture media. Effects on steroidogenesis was measured by real-time polymerase chain reaction (RT-PCR). Induction of apoptosis was evaluated by flow cytometry. RESULTS Combination Index (CI) for sHDL and either etoposide (E), cisplatin (P), or mitotane (M) demonstrated synergy (CI < 1) for antiproliferation. Alone or in combination with the chemotherapy drugs, sHDL was able to decrease cortisol production by 70-90% compared with P alone or controls (P < .01). RT-PCR indicated inhibition of steroidogenic enzymes for sHDL (P < .01 vs no sHDL). Combination therapy with sHDL increased apoptosis by 30-50% compared with drug or sHDL alone (P < .03), confirmed by a decrease in the mitochondrial potential. CONCLUSION sHDL can act synergistically and lessen the amount of M/E/P needed for anticancer efficacy in ACC in part owing to cholesterol starvation. This novel treatment strategy warrants further investigation translationally.
Collapse
|
64
|
Shannahan JH, Bai W, Brown JM. Implications of scavenger receptors in the safe development of nanotherapeutics. ACTA ACUST UNITED AC 2015; 2:e811. [PMID: 26005702 DOI: 10.14800/rci.811] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nanomaterials (NMs) are being utilized in a variety of biomedical applications including drug delivery, diagnostics, and therapeutic targeting. These applications are made possible due to the unique physicochemical properties that are exhibited at the nanoscale. To ensure safe development of NMs for clinical use, it is necessary to understand their interactions with cells and specifically cell surface receptors, which will facilitate either their toxicity and/or clinical function. Recently our research and others have investigated the role of scavenger receptors in mediating NM-cell interactions and responses. Scavenger receptors are expressed by a variety of cell types that are first to encounter NMs during clinical use such as macrophages and endothelial cells. Scavenger receptors are recognized to facilitate uptake of a wide variety of ligands ranging from foreign substances to endogenous lipids/proteins. While interaction of NMs with scavenger receptors may allow therapeutic targeting in some instances, it also presents a challenge for the stealth delivery of NMs and avoidance of the scavenging capability of this class of receptors. Due to their role in facilitating immune responses, scavenger receptor-mediated inflammation is also of concern following NM delivery. The research highlighted in this brief review intends to summarize our current understanding regarding the consequences of NM-scavenger receptor interactions.
Collapse
Affiliation(s)
- Jonathan H Shannahan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Wei Bai
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Jared M Brown
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| |
Collapse
|
65
|
Yuan B, Wu C, Wang X, Wang D, Liu H, Guo L, Li XA, Han J, Feng H. High scavenger receptor class B type I expression is related to tumor aggressiveness and poor prognosis in breast cancer. Tumour Biol 2015; 37:3581-8. [PMID: 26456958 DOI: 10.1007/s13277-015-4141-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/25/2015] [Indexed: 01/25/2023] Open
Abstract
Scavenger receptor class B type I (SR-BI) has been linked to the development and progression of breast cancer. However, its clinical significance in breast cancer remains unclear. Here, we evaluated SR-BI expression in a well-characterized breast cancer tissue microarray by immunohistochemistry. High SR-BI expression was observed in 54 % of all breast cancer cases and was significantly associated with advanced pTNM stage (P = 0.002), larger tumor size (P = 0.023), lymph node metastasis (P = 0.012), and the absence of ER (P = 0.014). The Kaplan-Meier survival analysis revealed that patients with high SR-BI expression had significantly shorter overall survival (OS) (P = 0.004). Moreover, multivariate analysis with adjustment for other prognostic factors confirmed that SR-BI was an independent prognostic factor for patient outcome (P = 0.017). Overall, our study demonstrated that high SR-BI expression was related to conventional parameters indicative of more aggressive tumor type and may serve as a new prognostic marker for poor clinical outcome in human breast cancer.
Collapse
Affiliation(s)
- Baoying Yuan
- Cancer Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, People's Republic of China
| | - Changshun Wu
- Department of Orthopedics, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China
| | - Xingwen Wang
- Cancer Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, People's Republic of China
| | - Dan Wang
- Department of Pediatrics, Saha Cardiovascular Research Center, University of Kentucky College of Medicine, 741 S. Limestone Street, Lexington, KY, 40536, USA
| | - Huiling Liu
- Cancer Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, People's Republic of China
| | - Ling Guo
- Department of Central Lab, Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, People's Republic of China
| | - Xiang-An Li
- Department of Pediatrics, Saha Cardiovascular Research Center, University of Kentucky College of Medicine, 741 S. Limestone Street, Lexington, KY, 40536, USA.
| | - Junqing Han
- Cancer Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, People's Republic of China.
| | - Hong Feng
- Cancer Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, People's Republic of China.
| |
Collapse
|
66
|
Schörghofer D, Kinslechner K, Preitschopf A, Schütz B, Röhrl C, Hengstschläger M, Stangl H, Mikula M. The HDL receptor SR-BI is associated with human prostate cancer progression and plays a possible role in establishing androgen independence. Reprod Biol Endocrinol 2015; 13:88. [PMID: 26251134 PMCID: PMC4528807 DOI: 10.1186/s12958-015-0087-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/31/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Human prostate cancer represents one of the most frequently diagnosed cancers in men worldwide. Currently, diagnostic methods are insufficient to identify patients at risk for aggressive prostate cancer, which is essential for early treatment. Recent data indicate that elevated cholesterol levels in the plasma are a prerequisite for the progression of prostate cancer. Here, we analyzed clinical prostate cancer samples for the expression of receptors involved in cellular cholesterol uptake. METHODS We screened mRNA microarray files of prostate cancer samples for alterations in the expression levels of cholesterol transporters. Furthermore, we performed immunohistochemistry analysis on human primary prostate cancer tissue sections derived from patients to investigate the correlation of SR-BI with clinicopathological parameters and the mTOR target pS6. RESULTS In contrast to LDLR, we identified SR-BI mRNA and protein expression to be induced in high Gleason grade primary prostate cancers. Histologic analysis of prostate biopsies revealed that 53.6 % of all cancer samples and none of the non-cancer samples showed high SR-BI staining intensity. The disease-free survival time was reduced (P = 0.02) in patients expressing high intra-tumor levels of SR-BI. SR-BI mRNA correlated with HSD17B1 and HSD3B1 and SR-BI protein staining showed correlation with active ribosomal protein S6 (RS = 0.828, P < 0.00001). CONCLUSIONS We identified SR-BI to indicate human prostate cancer formation, suggesting that increased levels of SR-BI may be involved in the generation of a castration-resistant phenotype.
Collapse
Affiliation(s)
- David Schörghofer
- Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| | - Katharina Kinslechner
- Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| | - Andrea Preitschopf
- Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| | - Birgit Schütz
- Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| | - Clemens Röhrl
- Institute of Medical Chemistry, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| | - Markus Hengstschläger
- Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| | - Herbert Stangl
- Institute of Medical Chemistry, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| | - Mario Mikula
- Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| |
Collapse
|
67
|
Abstract
Scavenger receptors constitute a large family of evolutionally conserved protein molecules that are structurally and functionally diverse. Although scavenger receptors were originally identified based on their capacity to scavenge modified lipoproteins, these molecules have been shown to recognize and bind to a broad spectrum of ligands, including modified and unmodified host-derived molecules or microbial components. As a major subset of innate pattern recognition receptors, scavenger receptors are mainly expressed on myeloid cells and function in a wide range of biological processes, such as endocytosis, adhesion, lipid transport, antigen presentation, and pathogen clearance. In addition to playing a crucial role in maintenance of host homeostasis, scavenger receptors have been implicated in the pathogenesis of a number of diseases, e.g., atherosclerosis, neurodegeneration, or metabolic disorders. Emerging evidence has begun to reveal these receptor molecules as important regulators of tumor behavior and host immune responses to cancer. This review summarizes our current understanding on the newly identified, distinct functions of scavenger receptors in cancer biology and immunology. The potential of scavenger receptors as diagnostic biomarkers and novel targets for therapeutic interventions to treat malignancies is also highlighted.
Collapse
Affiliation(s)
- Xiaofei Yu
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - John R Subjeck
- Department of Cellular Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA.
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|
68
|
Huang H, Cruz W, Chen J, Zheng G. Learning from biology: synthetic lipoproteins for drug delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 7:298-314. [PMID: 25346461 PMCID: PMC4397116 DOI: 10.1002/wnan.1308] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/22/2014] [Accepted: 09/02/2014] [Indexed: 12/15/2022]
Abstract
Synthetic lipoproteins represent a relevant tool for targeted delivery of biological/chemical agents (chemotherapeutics, siRNAs, photosensitizers, and imaging contrast agents) into various cell types. These nanoparticles offer a number of advantages for drugs delivery over their native counterparts while retaining their natural characteristics and biological functions. Their ultra-small size (<30 nm), high biocompatibility, favorable circulation half-life, and natural ability to bind specific lipoprotein receptors, i.e., low-density lipoprotein receptor (LDLR) and Scavenger receptor class B member 1 (SRB1) that are found in a number of pathological conditions (e.g., cancer, atherosclerosis), make them superior delivery strategies when compared with other nanoparticle systems. We review the various approaches that have been developed for the generation of synthetic lipoproteins and their respective applications in vitro and in vivo. More specifically, we summarize the approaches employed to address the limitation on use of reconstituted lipoproteins by means of natural or recombinant apolipoproteins, as well as apolipoprotein mimetic molecules. Finally, we provide an overview of the advantages and disadvantages of these approaches and discuss future perspectives for clinical translation of these nanoparticles.
Collapse
Affiliation(s)
- Huang Huang
- DLVR Therapeutics Inc., Toronto, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada M5G 1L7
| | - William Cruz
- DLVR Therapeutics Inc., Toronto, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada M5G 1L7
| | - Juan Chen
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada M5G 1L7
| | - Gang Zheng
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada M5G 1L7
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada M5G 1L7
| |
Collapse
|
69
|
Dai L, Zhang Q, Li J, Shen X, Mu C, Cai K. Dendrimerlike mesoporous silica nanoparticles as pH-responsive nanocontainers for targeted drug delivery and bioimaging. ACS APPLIED MATERIALS & INTERFACES 2015; 7:7357-72. [PMID: 25765172 DOI: 10.1021/acsami.5b00746] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
In this work, we employed dendrimerlike mesoporous silica nanoparticles with hierarchical pores (HPSNs) to fabricate drug delivery system bioimaging and targeted tumor therapy in vivo. N,N-phenylenebis(salicylideneimine)dicarboxylic acid (Salphdc) was used both as the gatekeeper of HPSNs via pH-responsive coordination bonds between -COOH of Salphdc and In(3+) ions and as a fluorescence imaging agent. Folic acid was then conjugated to Salphdc as the targeting unit. The results revealed that the system could deliver model drug DOX to the tumor site with high efficiency and then cause cell apoptosis and tumor growth inhibition. Moreover, the conjugated Salphdc was proved to be a promising fluorescence probe for tracing distribution of the system in vivo. The study affords a potential nanoconainer for cancer therapy and biological imaging.
Collapse
Affiliation(s)
- Liangliang Dai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China
| | - Qingfeng Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China
| | - Jinghua Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China
| | - Xinkun Shen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China
| | - Caiyun Mu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
70
|
McMahon KM, Foit L, Angeloni NL, Giles FJ, Gordon LI, Thaxton CS. Synthetic high-density lipoprotein-like nanoparticles as cancer therapy. Cancer Treat Res 2015; 166:129-50. [PMID: 25895867 PMCID: PMC4418545 DOI: 10.1007/978-3-319-16555-4_6] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
High-density lipoproteins (HDL) are diverse natural nanoparticles that carry cholesterol and are best known for the role that they play in cardiovascular disease. However, due to their unique targeting capabilities, diverse molecular cargo, and natural functions beyond cholesterol transport, it is becoming increasingly appreciated that HDLs are critical to cancer development and progression. Accordingly, this chapter highlights ongoing research focused on the connections between HDL and cancer in order to design new drugs and targeted drug delivery vehicles. Research is focused on synthesizing biomimetic HDL-like nanoparticles (NP) that can be loaded with diverse therapeutic cargo (e.g., chemotherapies, nucleic acids, proteins) and specifically targeted to cancer cells. Beyond drug delivery, new data is emerging that HDL-like NPs may be therapeutically active in certain tumor types, for example, B cell lymphoma. Overall, HDL-like NPs are becoming increasingly appreciated as targeted, biocompatible, and efficient therapies for cancer, and may soon become indispensable agents in the cancer therapeutic armamentarium.
Collapse
Affiliation(s)
- Kaylin M. McMahon
- Northwestern University, Feinberg School of Medicine, Department of Urology, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611 United States
- Simpson Querrey Institute (SQI), 303 E. Superior St, Chicago, IL 60611 United States
| | - Linda Foit
- Northwestern University, Feinberg School of Medicine, Department of Urology, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611 United States
- Simpson Querrey Institute (SQI), 303 E. Superior St, Chicago, IL 60611 United States
| | - Nicholas L. Angeloni
- Northwestern University, Feinberg School of Medicine, Department of Urology, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611 United States
- Simpson Querrey Institute (SQI), 303 E. Superior St, Chicago, IL 60611 United States
| | - Francis J. Giles
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, 645 N. Michigan Ave, Chicago, IL 60611, USA
| | - Leo I. Gordon
- Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611
| | - C. Shad Thaxton
- Northwestern University, Feinberg School of Medicine, Department of Urology, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611 United States
- Simpson Querrey Institute (SQI), 303 E. Superior St, Chicago, IL 60611 United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611
- International Institute for Nanotechnology (IIN), Northwestern University, 2145 Sheridan Rd. Evanston IL. 60208, United States
- Corresponding Author:
| |
Collapse
|
71
|
Savolainen MJ. Epidemiology: disease associations and modulators of HDL-related biomarkers. Handb Exp Pharmacol 2015; 224:259-283. [PMID: 25522991 DOI: 10.1007/978-3-319-09665-0_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Epidemiological studies have shown an inverse association between high-density lipoprotein cholesterol (HDL-C) levels and risk of ischemic heart disease. In addition, a low level of HDL-C has been shown to be a risk factor for other diseases not related to atherosclerosis. However, recent studies have not supported a causal effect of HDL-C in the development of atherosclerosis. Furthermore, new drugs markedly elevating HDL-C levels have been disappointing with respect to clinical endpoints. Earlier, most studies have focused almost exclusively on the total HDL-C without regard to the chemical composition or multiple subclasses of HDL particles. Recently, there have been efforts to dissect the HDL fraction into as many well-defined subfractions and individual molecules of HDL particles as possible. On the other hand, the focus is shifting from the structure and composition to the function of HDL particles. Biomarkers and mechanisms that could potentially explain the beneficial characteristics of HDL particles unrelated to their cholesterol content have been sought with sophisticated methods such as proteomics, lipidomics, metabonomics, and function studies including efflux capacity. These new approaches have been used in order to resolve the complex effects of diseases, conditions, environmental factors, and genes in relation to the protective role of HDL but high-throughput methods are still needed for large-scale epidemiological studies.
Collapse
Affiliation(s)
- Markku J Savolainen
- Department of Internal Medicine, Institute of Clinical Medicine, University of Oulu, Kajaanintie 50, 5000, 90014, Oulu, Finland,
| |
Collapse
|
72
|
Foit L, Giles FJ, Gordon LI, Thaxton CS. Synthetic high-density lipoprotein-like nanoparticles for cancer therapy. Expert Rev Anticancer Ther 2014; 15:27-34. [PMID: 25487833 DOI: 10.1586/14737140.2015.990889] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
High-density lipoproteins (HDLs) are a diverse group of natural nanoparticles that are most well known for their role in cholesterol transport. However, HDLs have diverse functions that provide significant opportunities for cancer therapy. Presented is a focused review of the ways that synthetic versions of HDL have been used as targeted therapies for cancer, and as vehicles for the delivery of diverse therapeutic cargo to cancer cells. As such, synthetic HDLs are likely to play a central role in the development of next-generation cancer therapies.
Collapse
Affiliation(s)
- Linda Foit
- Department of Urology, Feinberg School of Medicine, Northwestern University, Tarry 16-703, 303 E. Chicago Ave. Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
73
|
Sun Y, Kim HS, Park J, Li M, Tian L, Choi Y, Choi BI, Jon S, Moon WK. MRI of breast tumor initiating cells using the extra domain-B of fibronectin targeting nanoparticles. Theranostics 2014; 4:845-57. [PMID: 24955145 PMCID: PMC4063982 DOI: 10.7150/thno.8343] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/19/2014] [Indexed: 01/16/2023] Open
Abstract
The identification of breast tumor initiating cells (BTICs) is important for the diagnosis and therapy of breast cancers. This study was undertaken to evaluate whether the extra domain-B of fibronectin (EDB-FN) could be used as a new biomarker for BTICs and whether EDB-FN targeting superparamagnetic iron oxide nanoparticles (SPIONs) could be used as a magnetic resonance imaging (MRI) contrast agent for BTIC imaging in vitro and in vivo. BTICs (NDY-1) exhibited high EDB-FN expression, whereas non-BTICs (MCF-7, BT-474, SUM-225, MDA-MB-231) did not exhibit EDB-FN expression. Furthermore, Cy3.3-labeled EDB-FN specific peptides (APTEDB) showed preferential binding to the targeted NDY-1 cells. To construct an EDB-FN targeted imaging probe, APTEDB was covalently attached to a thermally cross-linked SPION (TCL-SPION) to yield APTEDB-TCL-SPION. In the in vitro MRI of cell phantoms, selective binding of APTEDB-TCL-SPION to NDY-1 cells was evident, but little binding was observed in MCF-7 cells. After the intravenous injection of APTEDB-TCL-SPION into the NDY-1 mouse tumor xenograft model, a significant decrease in the signal within the tumor was observed in the T2*-weighted images; however, there was only a marginal change in the signal of non-targeting SPIONs such as APTscramble-TCL-SPION or TCL-SPION. Taken together, we report for the first time that EDB-FN was abundantly expressed in BTICs and may therefore be useful as a new biomarker for identifying BTICs. Our study also suggests that APTEDB-TCL-SPION could be used as an MRI contrast agent for BTIC imaging.
Collapse
Affiliation(s)
- Yujin Sun
- 1. Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
- 2. Department of Radiology, Yanbian University Hospital, 1327 JuZi Street, Yanji City, JiLin Province 133000, China
| | - Hoe Suk Kim
- 1. Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
| | - Jinho Park
- 3. KAIST Institute for the BioCentury, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Korea
| | - Mulan Li
- 1. Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
| | - Lianji Tian
- 1. Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
| | - YoonSeok Choi
- 1. Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
| | - Byung Ihn Choi
- 1. Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
| | - Sangyong Jon
- 3. KAIST Institute for the BioCentury, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Korea
| | - Woo Kyung Moon
- 1. Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
| |
Collapse
|
74
|
Luo H, Lu L, Yang F, Wang L, Yang X, Luo Q, Zhang Z. Nasopharyngeal cancer-specific therapy based on fusion peptide-functionalized lipid nanoparticles. ACS NANO 2014; 8:4334-4347. [PMID: 24766601 DOI: 10.1021/nn405989n] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Current treatment of advanced-stage nasopharyngeal carcinoma (NPC) is not satisfactory. Targeted therapies offer hope for extending survival. Here, we developed simple, robust, and NPC-specific therapeutic lipid nanoparticles based on a fusion peptide, α-NTP, made up of an amphipathic α-helical peptide (α-peptide) linked to an NPC-specific therapeutic peptide (NTP). We found that α-NTP not only retained the sub-30 nm nanostructure-controlling ability of the α-peptide but also displayed the enhanced NPC-targeting ability of the NTP, in which the α-peptide accelerated the uptake of the NTP by NPC cells, with a 4.8-fold increase. Following uptake, α-NTP-based lipid nanoparticles (α-NTP-LNs) exerted coordinated cytotoxicity by inducing cell death via apoptosis and autophagy. In vivo and ex vivo optical imaging data showed that systemically administered α-NTP-LNs efficiently accumulated in the NPC xenograft tumor and displayed high contrast between tumor and normal tissues, which was further confirmed by flow cytometry that there had been a 13-fold uptake difference between tumor cells and hepatocytes. More importantly, the therapeutic efficacy of α-NTP-LNs was specific to NPC xenograft formed with 5-8F cells but not to fibrosarcoma xenograft formed with HT1080 cells in vivo. The growth of 5-8F tumors was significantly inhibited by α-NTP-LNs, with more than 85% inhibition relative to control groups (e.g., α-NTP and PBS treatment). In a lung metastasis model of NPC, survival was significantly improved by α-NTP-LN treatment. In a word, these excellent properties of α-NTP-LNs worked in sync and synergistically, maximizing the therapeutic efficacy of α-NTP-LNs against NPC and its metastasis.
Collapse
Affiliation(s)
- Haiming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, and ‡MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology , Wuhan 430074, China
| | | | | | | | | | | | | |
Collapse
|
75
|
Luo M, Zhang Z, Li H, Qiao S, Liu Z, Fu L, Shen G, Luo Q. Multi-scale optical imaging of the delayed type hypersensitivity reaction attenuated by rapamycin. Am J Cancer Res 2014; 4:201-14. [PMID: 24465276 PMCID: PMC3900803 DOI: 10.7150/thno.7570] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 12/07/2013] [Indexed: 12/05/2022] Open
Abstract
Neutrophils and monocytes/macrophages (MMs) play important roles in the development of cell-mediated delayed type hypersensitivity (DTH). However, the dynamics of neutrophils and MMs during the DTH reaction and how the immunosuppressant rapamycin modulates their behavior in vivo are rarely reported. Here, we take advantage of multi-scale optical imaging techniques and a footpad DTH reaction model to non-invasively investigate the dynamic behavior and properties of immune cells from the whole field of the footpad to the cellular level. During the classic elicitation phase of the DTH reaction, both neutrophils and MMs obviously accumulated at inflammatory foci at 24 h post-challenge. Rapamycin treatment resulted in advanced neutrophil recruitment and vascular hyperpermeability at an early stage (4 h), the reduced accumulation of neutrophils (> 50% inhibition ratio) at 48 h, and the delayed involvement of MMs in inflammatory foci. The motility parameters of immune cells in the rapamycin-treated reaction at 4 h post-challenge displayed similar mean velocities, arrest durations, mean displacements, and confinements as the classic DTH reaction at 24 h. These results indicate that rapamycin treatment shortened the initial preparation stage of the DTH reaction and attenuated its intensity, which may be due to the involvement of T helper type 2 cells or regulatory T cells.
Collapse
|
76
|
Huang C, Jin H, Qian Y, Qi S, Luo H, Luo Q, Zhang Z. Hybrid melittin cytolytic Peptide-driven ultrasmall lipid nanoparticles block melanoma growth in vivo. ACS NANO 2013; 7:5791-5800. [PMID: 23790040 DOI: 10.1021/nn400683s] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The cytolytic peptide melittin is a potential anticancer candidate that may be able to overcome tumor drug resistance due to its lytic properties. However, in vivo applications of melittin are limited due to its main side effect, hemolysis, which is especially pronounced following intravenous administration. Here, we designed a hybrid cytolytic peptide, α-melittin, in which the N-terminus of melittin is linked to the C-terminus of an amphipathic α-helical peptide (α-peptide) via a GSG linker. The strong α-helical configuration allows α-melittin to interact with phospholipids and self-assemble into lipid nanoparticles, with a high efficiency for α-melittin encapsulation (>80%) and a strong ability to control the structure of the nanoparticle (~20 nm). This α-melittin-based lipid nanoparticle (α-melittin-NP) efficiently shields the positive charge of melittin (18.70 ± 0.90 mV) within the phospholipid monolayer, resulting in the generation of a neutral nanoparticle (2.45 ± 0.56 mV) with reduced cytotoxicity and a widened safe dosage range. Confocal imaging data confirmed that α-melittin peptides were efficiently released from the nanoparticles and were cytotoxic to the melanoma cells. Finally, α-melittin-NPs were administered to melanoma-bearing mice via intravenous injection. The growth of the melanoma cells was blocked by the α-melittin-NPs, with an 82.8% inhibition rate relative to the PBS-treated control group. No side effects of treatment were found in this study. Thus, the excellent properties of α-melittin-NP give it potential clinical applications in solid tumor therapeutics through intravenous administration.
Collapse
Affiliation(s)
- Chuan Huang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
| | | | | | | | | | | | | |
Collapse
|