1
|
Wen X, Zhang C, Tian Y, Miao Y, Liu S, Xu JJ, Ye D, He J. Smart Molecular Imaging and Theranostic Probes by Enzymatic Molecular In Situ Self-Assembly. JACS AU 2024; 4:2426-2450. [PMID: 39055152 PMCID: PMC11267545 DOI: 10.1021/jacsau.4c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Enzymatic molecular in situ self-assembly (E-MISA) that enables the synthesis of high-order nanostructures from synthetic small molecules inside a living subject has emerged as a promising strategy for molecular imaging and theranostics. This strategy leverages the catalytic activity of an enzyme to trigger probe substrate conversion and assembly in situ, permitting prolonging retention and congregating many molecules of probes in the targeted cells or tissues. Enhanced imaging signals or therapeutic functions can be achieved by responding to a specific enzyme. This E-MISA strategy has been successfully applied for the development of enzyme-activated smart molecular imaging or theranostic probes for in vivo applications. In this Perspective, we discuss the general principle of controlling in situ self-assembly of synthetic small molecules by an enzyme and then discuss the applications for the construction of "smart" imaging and theranostic probes against cancers and bacteria. Finally, we discuss the current challenges and perspectives in utilizing the E-MISA strategy for disease diagnoses and therapies, particularly for clinical translation.
Collapse
Affiliation(s)
- Xidan Wen
- Department
of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing 210008, China
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Chao Zhang
- Department
of Neurosurgery, Zhujiang Hospital, Southern
Medical University, Guangzhou 510282, China
| | - Yuyang Tian
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Yinxing Miao
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Shaohai Liu
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Jing-Juan Xu
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Deju Ye
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Jian He
- Department
of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing 210008, China
| |
Collapse
|
2
|
Tan S, Chen Z, Mironchik Y, Mori N, Penet MF, Si G, Krishnamachary B, Bhujwalla ZM. VEGF Overexpression Significantly Increases Nanoparticle-Mediated siRNA Delivery and Target-Gene Downregulation. Pharmaceutics 2022; 14:pharmaceutics14061260. [PMID: 35745832 PMCID: PMC9229257 DOI: 10.3390/pharmaceutics14061260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 02/01/2023] Open
Abstract
The availability of nanoparticles (NPs) to deliver small interfering RNA (siRNA) has significantly expanded the specificity and range of ‘druggable’ targets for precision medicine in cancer. This is especially important for cancers such as triple negative breast cancer (TNBC) for which there are no targeted treatments. Our purpose here was to understand the role of tumor vasculature and vascular endothelial growth factor (VEGF) overexpression in a TNBC xenograft in improving the delivery and function of siRNA NPs using in vivo as well as ex vivo imaging. We used triple negative MDA-MB-231 human breast cancer xenografts derived from cells engineered to overexpress VEGF to understand the role of VEGF and vascularization in NP delivery and function. We used polyethylene glycol (PEG) conjugated polyethylenimine (PEI) NPs to deliver siRNA that downregulates choline kinase alpha (Chkα), an enzyme that is associated with malignant transformation and tumor progression. Because Chkα converts choline to phosphocholine, effective delivery of Chkα siRNA NPs resulted in functional changes of a significant decrease in phosphocholine and total choline that was detected with 1H magnetic resonance spectroscopy (MRS). We observed a significant increase in NP delivery and a significant decrease in Chkα and phosphocholine in VEGF overexpressing xenografts. Our results demonstrated the importance of tumor vascularization in achieving effective siRNA delivery and downregulation of the target gene Chkα and its function.
Collapse
Affiliation(s)
- Shanshan Tan
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Zhihang Chen
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Noriko Mori
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
| | - Ge Si
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Zaver M. Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Correspondence:
| |
Collapse
|
3
|
Zhang Y, Cui H, Zhang R, Zhang H, Huang W. Nanoparticulation of Prodrug into Medicines for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101454. [PMID: 34323373 PMCID: PMC8456229 DOI: 10.1002/advs.202101454] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/16/2021] [Indexed: 05/28/2023]
Abstract
This article provides a broad spectrum about the nanoprodrug fabrication advances co-driven by prodrug and nanotechnology development to potentiate cancer treatment. The nanoprodrug inherits the features of both prodrug concept and nanomedicine know-how, attempts to solve underexploited challenge in cancer treatment cooperatively. Prodrugs can release bioactive drugs on-demand at specific sites to reduce systemic toxicity, this is done by using the special properties of the tumor microenvironment, such as pH value, glutathione concentration, and specific overexpressed enzymes; or by using exogenous stimulation, such as light, heat, and ultrasound. The nanotechnology, manipulating the matter within nanoscale, has high relevance to certain biological conditions, and has been widely utilized in cancer therapy. Together, the marriage of prodrug strategy which shield the side effects of parent drug and nanotechnology with pinpoint delivery capability has conceived highly camouflaged Trojan horse to maneuver cancerous threats.
Collapse
Affiliation(s)
- Yuezhou Zhang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| | - Huaguang Cui
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| | - Ruiqi Zhang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, FI-00520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-00520, Finland
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| |
Collapse
|
4
|
Pacheco-Torres J, Penet MF, Krishnamachary B, Mironchik Y, Chen Z, Bhujwalla ZM. PD-L1 siRNA Theranostics With a Dextran Nanoparticle Highlights the Importance of Nanoparticle Delivery for Effective Tumor PD-L1 Downregulation. Front Oncol 2021; 10:614365. [PMID: 33718115 PMCID: PMC7947807 DOI: 10.3389/fonc.2020.614365] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/30/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose The inhibition of immune checkpoints such as programmed cell death ligand-1 (PD-L1/CD274) with antibodies is providing novel opportunities to expose cancer cells to the immune system. Antibody based checkpoint blockade can, however, result in serious autoimmune complications because normal tissues also express immune checkpoints. As sequence-specific gene-silencing agents, the availability of siRNA has significantly expanded the specificity and range of “druggable” targets making them promising agents for precision medicine in cancer. Here, we have demonstrated the ability of a novel biodegradable dextran based theranostic nanoparticle (NP) to deliver siRNA downregulating PD-L1 in tumors. Optical imaging highlighted the importance of NP delivery and accumulation in tumors to achieve effective downregulation with siRNA NPs, and demonstrated low delivery and accumulation in several PD-L1 expressing normal tissues. Methods The dextran scaffold was functionalized with small molecules containing amine groups through acetal bonds. The NP was decorated with a Cy5.5 NIR probe allowing visualization of NP delivery, accumulation, and biodistribution. MDA-MB-231 triple negative human breast cancer cells were inoculated orthotopically or subcutaneously to achieve differences in vascular delivery in the tumors. Molecular characterization of PD-L1 mRNA and protein expression in cancer cells and tumors was performed with qRT-PCR and immunoblot analysis. Results The PD-L1 siRNA dextran NPs effectively downregulated PD-L1 in MDA-MB-231 cells. We identified a significant correlation between NP delivery and accumulation, and the extent of PD-L1 downregulation, with in vivo imaging. The size of the NP of ~ 20 nm allowed delivery through leaky tumor vasculature but not through the vasculature of high PD-L1 expressing normal tissue such as the spleen and lungs. Conclusions Here we have demonstrated, for the first time, the feasibility of downregulating PD-L1 in tumors using siRNA delivered with a biodegradable dextran polymer that was decorated with an imaging reporter. Our data demonstrate the importance of tumor NP delivery and accumulation in achieving effective downregulation, highlighting the importance of imaging in siRNA NP delivery. Effective delivery of these siRNA carrying NPs in the tumor but not in normal tissues may mitigate some of the side-effects of immune checkpoint inhibitors by sparing PD-L1 inhibition in these tissues.
Collapse
Affiliation(s)
- Jesus Pacheco-Torres
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhihang Chen
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Raikundalia S, Sa'Dom SAFM, Few LL, Too WCS. MicroRNA-367-3p induces apoptosis and suppresses migration of MCF-7 cells by downregulating the expression of human choline kinase α. Oncol Lett 2021; 21:183. [PMID: 33574922 PMCID: PMC7816280 DOI: 10.3892/ol.2021.12444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Choline kinase (ChK) catalyzes the first step in the CDP-choline pathway for the synthesis of phosphatidylcholine. The α isoform of this enzyme is overexpressed in various types of cancer and its inhibition or downregulation has been applied as an anticancer strategy. In spite of increasing attention being paid to ChK expression, as well as its activity and inhibition in cancer, there are only limited studies available on the regulation of ChK, including its regulation by microRNAs (miRNAs/miRs). The dysregulation of gene expression by miRNAs is a common cause for carcinogenesis. In the present study, miR-367-3p was predicted to target the 3′-untranslated region (UTR) of the ChK α (chka) mRNA transcript. The binding of miR-367-3p to the 3′-UTR of chka was validated by a luciferase assay. The effects of the miR-367-3p mimic on chka gene and protein expression levels were determined by reverse transcription-quantitative polymerase chain reaction and western blot analysis, respectively. miR-367-3p significantly downregulated the expression of chka to ~60% of the negative control. Cells transfected with miR-367-3p exhibited higher levels of apoptosis and a lower cell migration compared with the control. To the best of our knowledge, the present study provided the first experimental evidence of the regulation of chka expression by miR-367-3p. The pro-apoptotic and suppressive effects of miR-367-3p on cell migration were similar to the anticancer effects resulting from the inhibition of ChK enzyme activity or the knockdown of chka gene expression by small interfering RNA. Therefore, these findings may potentially lead to the use of miR-367-3p in anticancer strategies that target ChK.
Collapse
Affiliation(s)
- Sweta Raikundalia
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| | | | - Ling Ling Few
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| | - Wei Cun See Too
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| |
Collapse
|
6
|
Krishnamachary B, Mironchik Y, Jacob D, Goggins E, Kakkad S, Ofori F, Dore-Savard L, Bharti SK, Wildes F, Penet MF, Black ME, Bhujwalla ZM. Hypoxia theranostics of a human prostate cancer xenograft and the resulting effects on the tumor microenvironment. Neoplasia 2020; 22:679-688. [PMID: 33142234 PMCID: PMC7586064 DOI: 10.1016/j.neo.2020.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 12/22/2022] Open
Abstract
Developed a hypoxia theranostic imaging strategy to eliminate hypoxic cells. Hypoxic cell elimination resulted in fewer cancer associated fibroblasts (CAFs) Collagen 1 fiber patterns were altered with hypoxic cell elimination. cDNA nanoparticles with HRE driven prodrug enzyme expression can target hypoxia.
Hypoxia is frequently observed in human prostate cancer, and is associated with chemoresistance, radioresistance, metastasis, and castrate-resistance. Our purpose in these studies was to perform hypoxia theranostics by combining in vivo hypoxia imaging and hypoxic cancer cell targeting in a human prostate cancer xenograft. This was achieved by engineering PC3 human prostate cancer cells to express luciferase as well as a prodrug enzyme, yeast cytosine deaminase, under control of hypoxic response elements (HREs). Cancer cells display an adaptive response to hypoxia through the activation of several genes mediated by the binding of hypoxia inducible factors (HIFs) to HRE in the promoter region of target gene that results in their increased transcription. HIFs promote key steps in tumorigenesis, including angiogenesis, metabolism, proliferation, metastasis, and differentiation. HRE-driven luciferase expression allowed us to detect hypoxia in vivo to time the administration of the nontoxic prodrug 5-fluorocytosine that was converted by yeast cytosine deaminase, expressed under HRE regulation, to the chemotherapy agent 5-fluorouracil to target hypoxic cells. Conversion of 5-fluorocytosine to 5-fluorouracil was detected in vivo by 19F magnetic resonance spectroscopy. Morphological and immunohistochemical staining and molecular analyses were performed to characterize tumor microenvironment changes in cancer-associated fibroblasts, cell viability, collagen 1 fiber patterns, and HIF-1α. These studies expand our understanding of the effects of eliminating hypoxic cancer cells on the tumor microenvironment and in reducing stromal cell populations such as cancer-associated fibroblasts.
Collapse
Affiliation(s)
- Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Desmond Jacob
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eibhlin Goggins
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Samata Kakkad
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Francis Ofori
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Louis Dore-Savard
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Santosh Kumar Bharti
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Flonne Wildes
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Margaret E Black
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD; Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
7
|
Wang X, Rong G, Yan J, Pan D, Wang L, Xu Y, Yang M, Cheng Y. In Vivo Tracking of Fluorinated Polypeptide Gene Carriers by Positron Emission Tomography Imaging. ACS APPLIED MATERIALS & INTERFACES 2020; 12:45763-45771. [PMID: 32940028 DOI: 10.1021/acsami.0c11967] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Fluorinated polymers have attracted increasing attention in gene delivery and cytosolic protein delivery in recent years. In vivo tracking of fluorinated polymers will be of great importance to evaluate their biodistribution, clearance, and safety. However, tracking of polymeric carriers without changing their chemical structures remains a huge challenge. Herein, we reported a series of fluorinated poly-l-(lysine) (F-PLL) with high gene transfection efficiency and excellent biodegradation. Radionuclide 18F was radiolabeled on F-PLL by halogen replacement without chemical modification. The radiolabeling of F-PLL offers positron emission tomography (PET) imaging for in vivo tracking of the polymers. The biodistribution of F-PLL and the DNA complexes revealed by micro-PET imaging illustrated the rapid clearance of fluorinated polymers from liver and intestine after intravenous administration. The results demonstrated that the polymer F-PLL will not be accumulated in the liver and spleen when administrated as a gene carrier. This work presents a new strategy for in vivo tracking fluorinated polymers via PET imaging.
Collapse
Affiliation(s)
- Xinyu Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine. Wuxi 214063, China
| | - Guangyu Rong
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Junjie Yan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine. Wuxi 214063, China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine. Wuxi 214063, China
| | - Lizhen Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine. Wuxi 214063, China
| | - Yuping Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine. Wuxi 214063, China
| | - Min Yang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine. Wuxi 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
8
|
Serrano-Sevilla I, Artiga Á, Mitchell SG, De Matteis L, de la Fuente JM. Natural Polysaccharides for siRNA Delivery: Nanocarriers Based on Chitosan, Hyaluronic Acid, and Their Derivatives. Molecules 2019; 24:E2570. [PMID: 31311176 PMCID: PMC6680562 DOI: 10.3390/molecules24142570] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022] Open
Abstract
Natural polysaccharides are frequently used in the design of drug delivery systems due to their biocompatibility, biodegradability, and low toxicity. Moreover, they are diverse in structure, size, and charge, and their chemical functional groups can be easily modified to match the needs of the final application and mode of administration. This review focuses on polysaccharidic nanocarriers based on chitosan and hyaluronic acid for small interfering RNA (siRNA) delivery, which are highly positively and negatively charged, respectively. The key properties, strengths, and drawbacks of each polysaccharide are discussed. In addition, their use as efficient nanodelivery systems for gene silencing applications is put into context using the most recent examples from the literature. The latest advances in this field illustrate effectively how chitosan and hyaluronic acid can be modified or associated with other molecules in order to overcome their limitations to produce optimized siRNA delivery systems with promising in vitro and in vivo results.
Collapse
Affiliation(s)
- Inés Serrano-Sevilla
- Instituto de Ciencia de Materiales de Aragón (ICMA), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
- CIBER-BBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Álvaro Artiga
- Instituto de Ciencia de Materiales de Aragón (ICMA), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
- CIBER-BBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Scott G Mitchell
- Instituto de Ciencia de Materiales de Aragón (ICMA), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
- CIBER-BBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura De Matteis
- CIBER-BBN, Instituto de Salud Carlos III, Madrid, Spain.
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza, C/Mariano Esquillor s/n, 50018 Zaragoza, Spain.
| | - Jesús M de la Fuente
- Instituto de Ciencia de Materiales de Aragón (ICMA), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain.
- CIBER-BBN, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
9
|
Bhujwalla ZM, Kakkad S, Chen Z, Jin J, Hapuarachchige S, Artemov D, Penet MF. Theranostics and metabolotheranostics for precision medicine in oncology. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2018; 291:141-151. [PMID: 29705040 PMCID: PMC5943142 DOI: 10.1016/j.jmr.2018.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/12/2018] [Accepted: 03/07/2018] [Indexed: 05/14/2023]
Abstract
Most diseases, especially cancer, would significantly benefit from precision medicine where treatment is shaped for the individual. The concept of theragnostics or theranostics emerged around 2002 to describe the incorporation of diagnostic assays into the selection of therapy for this purpose. Increasingly, theranostics has been used for strategies that combine noninvasive imaging-based diagnostics with therapy. Within the past decade theranostic imaging has transformed into a rapidly expanding field that is located at the interface of diagnosis and therapy. A critical need in cancer treatment is to minimize damage to normal tissue. Molecular imaging can be applied to identify targets specific to cancer with imaging, design agents against these targets to visualize their delivery, and monitor response to treatment, with the overall purpose of minimizing collateral damage. Genomic and proteomic profiling can provide an extensive 'fingerprint' of each tumor. With this cancer fingerprint, theranostic agents can be designed to personalize treatment for precision medicine of cancer, and minimize damage to normal tissue. Here, for the first time, we have introduced the term 'metabolotheranostics' to describe strategies where disease-based alterations in metabolic pathways detected by MRS are specifically targeted with image-guided delivery platforms to achieve disease-specific therapy. The versatility of MRI and MRS in molecular and functional imaging makes these technologies especially important in theranostic MRI and 'metabolotheranostics'. Our purpose here is to provide insights into the capabilities and applications of this exciting new field in cancer treatment with a focus on MRI and MRS.
Collapse
Affiliation(s)
- Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Samata Kakkad
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhihang Chen
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiefu Jin
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sudath Hapuarachchige
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dmitri Artemov
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
10
|
Chen Z, Krishnamachary B, Penet MF, Bhujwalla ZM. Acid-degradable Dextran as an Image Guided siRNA Carrier for COX-2 Downregulation. Am J Cancer Res 2018; 8:1-12. [PMID: 29290789 PMCID: PMC5743456 DOI: 10.7150/thno.21052] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/27/2017] [Indexed: 01/10/2023] Open
Abstract
Purpose: Effective in vivo delivery of siRNA to silence genes is a highly sought-after goal in the treatment of multiple diseases. Cyclooxygenase-2 (COX-2) is a major mediator of inflammation and its effective and specific downregulation has been of major interest to treat conditions ranging from auto-immune diseases to gastric inflammation and cancer. Here we developed a novel and efficient method to produce a multiple imaging reporter labeled cationic dextran nanopolymer with cleavable positive charge groups for COX-2 siRNA delivery. Methods: Small molecules containing amine groups were conjugated to the dextran scaffold through acetal bonds that were cleaved in weak acid conditions. With multiple imaging reporters located on different regions of the nanopolymer, cleavage of acetal bonds was visualized and quantified by imaging, for the first time, in cancer cells and tumors. Results: The biocompatibility of dextran and the rapid cleavage and release of amine groups minimized proinflammatory side effects and COX-2 induction observed with other siRNA carriers, to successfully achieve COX-2 downregulation in cancer cells and tumors. Imaging results confirmed that this nanoplex, consisting of the dextran nanopolymer with COX-2 siRNA, accumulated in tumors, and the amine functional groups were rapidly cleaved in cancer cells and tumors. Along with effective downregulation of COX-2, we also demonstrated, for the first time, effective downregulation of its major product prostaglandin E2 (PGE2). Conclusions: We successfully developed an efficient method to produce an acid-degradable dextran nanopolymer containing cleavable amine groups as the siRNA carrier. Because of its biocompatibility, this degradable dextran delivered COX-2 siRNA within tumors and efficiently downregulated COX-2 expression.
Collapse
|
11
|
Yuan MH, Wei LX, Zhou RS, Xu HF, Wang JY, Bai QR. Therapeutic effects of adenovirus-mediated CD and NIS expression combined with Na 131I/5-FC on human thyroid cancer. Oncol Lett 2017; 14:7431-7436. [PMID: 29344184 DOI: 10.3892/ol.2017.7175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 04/07/2017] [Indexed: 12/13/2022] Open
Abstract
Thyroid cancer is the most common type of malignant endocrine tumor diagnosed. Previous studies have indicated that gene therapy is the most promising and effective therapeutic method for thyroid cancer. Therefore, in the present study, Na131I/5-fluorocytosine (5-FC) treatment was combined with cytosine deaminase (CD, encoded by the CDA gene) and sodium iodide symporter (NIS, encoded by the SLC5A5 gene) to act together as a therapeutic tool for thyroid cancer. The present study explored the combined cytotoxic effects of adenovirus-mediated CD and NIS under the control of the progression elevated gene-3 (PEG-3) promoter (Ad-PEG-3-CD-NIS) with Na131I/5-FC against the human thyroid cancer TT cell line in vitro. The PEG-3 fragment was obtained by polymerase chain reaction (PCR) using rat genomic DNA as the template, and then Ad-PEG-3-CDA-SLC5A5 was constructed using XbaI. TT cells were transfected by recombinant adenovirus. The method of reverse transcription-quantitative PCR was performed to test the expression of CD and NIS at the level of transcription. The morphological change was assessed by fluorescence microscopy and investigated by western blot analysis. An MTT assay was used to determine the number of living cells inhibited by single or combination therapies on TT cells. The results indicated that the PEG-3 was successfully cloned, and was also positively regulated in 293 cells. CDA and SLC5A5 genes were highly expressed in TT cells. Na131I combined with 5-FC significantly decreased the human thyroid cancer cells. In conclusion, combination therapy of Ad-PEG3-CDA-SLC5A5 and Na131I/5-FC induces significantly more apoptotic characteristics than either single treatment with Ad-PEG-3-CDA-SLC5A5 or Na131I/5-FC, and low doses of Ad-PEG-3-CDA-SLC5A5 enhanced the cytotoxic effects.
Collapse
Affiliation(s)
- Meng-Hui Yuan
- Department of Nuclear Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Long-Xiao Wei
- Department of Nuclear Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Run-Suo Zhou
- Department of Nuclear Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Hai-Feng Xu
- Department of Nuclear Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Jun-Yan Wang
- Department of Nuclear Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Qian-Rong Bai
- Department of Nuclear Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
12
|
Genetically encoded iron-associated proteins as MRI reporters for molecular and cellular imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10. [DOI: 10.1002/wnan.1482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 04/18/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023]
|
13
|
Cheng M, Bhujwalla ZM, Glunde K. Targeting Phospholipid Metabolism in Cancer. Front Oncol 2016; 6:266. [PMID: 28083512 PMCID: PMC5187387 DOI: 10.3389/fonc.2016.00266] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022] Open
Abstract
All cancers tested so far display abnormal choline and ethanolamine phospholipid metabolism, which has been detected with numerous magnetic resonance spectroscopy (MRS) approaches in cells, animal models of cancer, as well as the tumors of cancer patients. Since the discovery of this metabolic hallmark of cancer, many studies have been performed to elucidate the molecular origins of deregulated choline metabolism, to identify targets for cancer treatment, and to develop MRS approaches that detect choline and ethanolamine compounds for clinical use in diagnosis and treatment monitoring. Several enzymes in choline, and recently also ethanolamine, phospholipid metabolism have been identified, and their evaluation has shown that they are involved in carcinogenesis and tumor progression. Several already established enzymes as well as a number of emerging enzymes in phospholipid metabolism can be used as treatment targets for anticancer therapy, either alone or in combination with other chemotherapeutic approaches. This review summarizes the current knowledge of established and relatively novel targets in phospholipid metabolism of cancer, covering choline kinase α, phosphatidylcholine-specific phospholipase D1, phosphatidylcholine-specific phospholipase C, sphingomyelinases, choline transporters, glycerophosphodiesterases, phosphatidylethanolamine N-methyltransferase, and ethanolamine kinase. These enzymes are discussed in terms of their roles in oncogenic transformation, tumor progression, and crucial cancer cell properties such as fast proliferation, migration, and invasion. Their potential as treatment targets are evaluated based on the current literature.
Collapse
Affiliation(s)
- Menglin Cheng
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Glunde
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Penet MF, Jin J, Chen Z, Bhujwalla ZM. Magnetic Resonance Imaging and Spectroscopy in Cancer Theranostic Imaging. Top Magn Reson Imaging 2016; 25:215-221. [PMID: 27748706 PMCID: PMC5893223 DOI: 10.1097/rmr.0000000000000098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
With its exquisite anatomical resolution and wide-ranging functional imaging capabilities, magnetic resonance imaging (MRI) has found multiple applications in detection, staging, and monitoring treatment response in cancer. The metabolic information provided by magnetic resonance spectroscopy (MRS) is being actively investigated to complement MRI parameters, as well as existing biomarkers, in cancer detection and in monitoring response to treatment. Located at the interface of detection and therapy, theranostic imaging is a rapidly expanding new field that is showing significant promise for precision medicine of cancer. Innovations in the development of novel nanoparticles decorated with imaging reporters that can be used to deliver therapeutic cargo to specific cells and environments have provided new roles for MRI and MRS in theranostic imaging.
Collapse
Affiliation(s)
- Marie-France Penet
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jiefu Jin
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zhihang Chen
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zaver M. Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
15
|
Wang J, Mi P, Lin G, Wáng YXJ, Liu G, Chen X. Imaging-guided delivery of RNAi for anticancer treatment. Adv Drug Deliv Rev 2016; 104:44-60. [PMID: 26805788 DOI: 10.1016/j.addr.2016.01.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 11/27/2015] [Accepted: 01/13/2016] [Indexed: 12/12/2022]
Abstract
The RNA interference (RNAi) technique is a new modality for cancer therapy, and several candidates are being tested clinically. In the development of RNAi-based therapeutics, imaging methods can provide a visible and quantitative way to investigate the therapeutic effect at anatomical, cellular, and molecular level; to noninvasively trace the distribution; to and study the biological processes in preclinical and clinical stages. Their abilities are important not only for therapeutic optimization and evaluation but also for shortening of the time of drug development to market. Typically, imaging-functionalized RNAi therapeutics delivery that combines nanovehicles and imaging techniques to study and improve their biodistribution and accumulation in tumor site has been progressively integrated into anticancer drug discovery and development processes. This review presents an overview of the current status of translating the RNAi cancer therapeutics in the clinic, a brief description of the biological barriers in drug delivery, and the roles of imaging in aspects of administration route, systemic circulation, and cellular barriers for the clinical translation of RNAi cancer therapeutics, and with partial content for discussing the safety concerns. Finally, we focus on imaging-guided delivery of RNAi therapeutics in preclinical development, including the basic principles of different imaging modalities, and their advantages and limitations for biological imaging. With growing number of RNAi therapeutics entering the clinic, various imaging methods will play an important role in facilitating the translation of RNAi cancer therapeutics from bench to bedside.
Collapse
|
16
|
Huang J, Li Y, Orza A, Lu Q, Guo P, Wang L, Yang L, Mao H. Magnetic Nanoparticle Facilitated Drug Delivery for Cancer Therapy with Targeted and Image-Guided Approaches. ADVANCED FUNCTIONAL MATERIALS 2016; 26:3818-3836. [PMID: 27790080 PMCID: PMC5077153 DOI: 10.1002/adfm.201504185] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
With rapid advances in nanomedicine, magnetic nanoparticles (MNPs) have emerged as a promising theranostic tool in biomedical applications, including diagnostic imaging, drug delivery and novel therapeutics. Significant preclinical and clinical research has explored their functionalization, targeted delivery, controllable drug release and image-guided capabilities. To further develop MNPs for theranostic applications and clinical translation in the future, we attempt to provide an overview of the recent advances in the development and application of MNPs for drug delivery, specifically focusing on the topics concerning the importance of biomarker targeting for personalized therapy and the unique magnetic and contrast-enhancing properties of theranostic MNPs that enable image-guided delivery. The common strategies and considerations to produce theranostic MNPs and incorporate payload drugs into MNP carriers are described. The notable examples are presented to demonstrate the advantages of MNPs in specific targeting and delivering under image guidance. Furthermore, current understanding of delivery mechanisms and challenges to achieve efficient therapeutic efficacy or diagnostic capability using MNP-based nanomedicine are discussed.
Collapse
Affiliation(s)
- Jing Huang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anamaria Orza
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Peng Guo
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, USA. Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Liya Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
17
|
Shahbazi R, Ozpolat B, Ulubayram K. Oligonucleotide-based theranostic nanoparticles in cancer therapy. Nanomedicine (Lond) 2016; 11:1287-308. [PMID: 27102380 DOI: 10.2217/nnm-2016-0035] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Theranostic approaches, combining the functionality of both therapy and imaging, have shown potential in cancer nanomedicine. Oligonucleotides such as small interfering RNA and microRNA, which are powerful therapeutic agents, have been effectively employed in theranostic systems against various cancers. Nanoparticles are used to deliver oligonucleotides into tumors by passive or active targeting while protecting the oligonucleotides from nucleases in the extracellular environment. The use of quantum dots, iron oxide nanoparticles and gold nanoparticles and tagging with contrast agents, like fluorescent dyes, optical or magnetic agents and various radioisotopes, has facilitated early detection of tumors and evaluation of therapeutic efficacy. In this article, we review the advantages of theranostic applications in cancer therapy and imaging, with special attention to oligonucleotide-based therapeutics.
Collapse
Affiliation(s)
- Reza Shahbazi
- Department of Nanotechnology & Nanomedicine, Institute for Graduate Studies in Science & Engineering, Hacettepe University, Ankara 06532, Turkey
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kezban Ulubayram
- Department of Nanotechnology & Nanomedicine, Institute for Graduate Studies in Science & Engineering, Hacettepe University, Ankara 06532, Turkey.,Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey.,Department of Bioengineering, Institute for Graduate Studies in Science & Engineering, Hacettepe University, Ankara 06532, Turkey
| |
Collapse
|
18
|
Chen Z, Krishnamachary B, Bhujwalla ZM. Degradable Dextran Nanopolymer as a Carrier for Choline Kinase (ChoK) siRNA Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2016; 6:E34. [PMID: 28344291 PMCID: PMC5302479 DOI: 10.3390/nano6020034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/19/2016] [Accepted: 02/04/2016] [Indexed: 01/28/2023]
Abstract
Although small interfering RNA (siRNA) therapy has proven to be a specific and effective treatment in cells, the delivery of siRNA is a challenge for the applications of siRNA therapy. We present a degradable dextran with amine groups as an siRNA nano-carrier. In our nano-carrier, the amine groups are conjugated to the dextran platform through the acetal bonds, which are acid sensitive. Therefore this siRNA carrier is stable in neutral and basic conditions, while the amine groups can be cleaved and released from dextran platform under weak acid conditions (such as in endosomes). The cleavage and release of amine groups can reduce the toxicity of cationic polymer and enhance the transfection efficiency. We successfully applied this nano-carrier to deliver choline kinase (ChoK) siRNA for ChoK inhibition in cells.
Collapse
Affiliation(s)
- Zhihang Chen
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Balaji Krishnamachary
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Zaver M Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
19
|
Abstract
Small interfering RNA (siRNA) is emerging as a class of therapeutic with extremely high potential, particularly in the field of oncology. Despite this growing interest, further understanding of how siRNA behaves in vivo is still required before significant uptake into clinical application. To this end, many molecular imaging modalities have been utilised to gain a better understanding of the biodistribution and pharmacokinetics of administered siRNA and delivery vehicles. This highlight aims to provide an overview of the current state of the field for preclinical imaging of siRNA delivery.
Collapse
|
20
|
Penet MF, Chen Z, Mori N, Krishnamachary B, Bhujwalla ZM. Magnetic Resonance Spectroscopy of siRNA-Based Cancer Therapy. Methods Mol Biol 2016; 1372:37-47. [PMID: 26530913 DOI: 10.1007/978-1-4939-3148-4_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Small interfering RNA (siRNA) is routinely used as a biological tool to silence specific genes, and is under active investigation in cancer treatment strategies. Noninvasive magnetic resonance spectroscopy (MRS) provides the ability to assess the functional effects of siRNA-mediated gene silencing in cultured cancer cells, and following nanoparticle-based delivery in tumors in vivo. Here we describe the use of siRNA to downregulate choline kinase, a critical enzyme in choline phospholipid metabolism of cancer cells and tumors, and the use of (1)H MRS of cells and (1)H magnetic resonance spectroscopic imaging (MRSI) of tumors to assess the efficacy of the downregulation.
Collapse
Affiliation(s)
- Marie-France Penet
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD, 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhihang Chen
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Noriko Mori
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Chen Z, Penet MF, Krishnamachary B, Banerjee SR, Pomper MG, Bhujwalla ZM. PSMA-specific theranostic nanoplex for combination of TRAIL gene and 5-FC prodrug therapy of prostate cancer. Biomaterials 2015; 80:57-67. [PMID: 26706476 DOI: 10.1016/j.biomaterials.2015.11.048] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/13/2015] [Accepted: 11/29/2015] [Indexed: 12/21/2022]
Abstract
Metastatic prostate cancer causes significant morbidity and mortality and there is a critical unmet need for effective treatments. We have developed a theranostic nanoplex platform for combined imaging and therapy of prostate cancer. Our prostate-specific membrane antigen (PSMA) targeted nanoplex is designed to deliver plasmid DNA encoding tumor necrosis factor related apoptosis-inducing ligand (TRAIL), together with bacterial cytosine deaminase (bCD) as a prodrug enzyme. Nanoplex specificity was tested using two variants of human PC3 prostate cancer cells in culture and in tumor xenografts, one with high PSMA expression and the other with negligible expression levels. The expression of EGFP-TRAIL was demonstrated by fluorescence optical imaging and real-time PCR. Noninvasive (19)F MR spectroscopy detected the conversion of the nontoxic prodrug 5-fluorocytosine (5-FC) to cytotoxic 5-fluorouracil (5-FU) by bCD. The combination strategy of TRAIL gene and 5-FC/bCD therapy showed significant inhibition of the growth of prostate cancer cells and tumors. These data demonstrate that the PSMA-specific theranostic nanoplex can deliver gene therapy and prodrug enzyme therapy concurrently for precision medicine in metastatic prostate cancer.
Collapse
Affiliation(s)
- Zhihang Chen
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, 21205, USA
| | - Marie-France Penet
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, 21205, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Balaji Krishnamachary
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, 21205, USA
| | - Sangeeta R Banerjee
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, 21205, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Martin G Pomper
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, 21205, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Zaver M Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, 21205, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
22
|
Penet MF, Krishnamachary B, Chen Z, Jin J, Bhujwalla ZM. Molecular imaging of the tumor microenvironment for precision medicine and theranostics. Adv Cancer Res 2015; 124:235-56. [PMID: 25287691 DOI: 10.1016/b978-0-12-411638-2.00007-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Morbidity and mortality from cancer and their associated conditions and treatments continue to extract a heavy social and economic global burden despite the transformative advances in science and technology in the twenty-first century. In fact, cancer incidence and mortality are expected to reach pandemic proportions by 2025, and costs of managing cancer will escalate to trillions of dollars. The inability to establish effective cancer treatments arises from the complexity of conditions that exist within tumors, the plasticity and adaptability of cancer cells coupled with their ability to escape immune surveillance, and the co-opted stromal cells and microenvironment that assist cancer cells in survival. Stromal cells, although destroyed together with cancer cells, have an ever-replenishing source that can assist in resurrecting tumors from any residual cancer cells that may survive treatment. The tumor microenvironment landscape is a continually changing landscape, with spatial and temporal heterogeneities that impact and influence cancer treatment outcome. Importantly, the changing landscape of the tumor microenvironment can be exploited for precision medicine and theranostics. Molecular and functional imaging can play important roles in shaping and selecting treatments to match this landscape. Our purpose in this review is to examine the roles of molecular and functional imaging, within the context of the tumor microenvironment, and the feasibility of their applications for precision medicine and theranostics in humans.
Collapse
Affiliation(s)
- Marie-France Penet
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Balaji Krishnamachary
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhihang Chen
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiefu Jin
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zaver M Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
23
|
Abstract
Viruses are promising vehicles that result in high gene expression level, but issues of safety and virulent nature prevented its extensive use. Therefore, nonviral approach was investigated with the intervention of nanomedicine. The science of nanomedicine offered an excellent platform for therapeutic delivery as they provide options to include functionalities and engineer the system. As the term 'nano' refers to the generation of a very small dimension structure, their unique physicochemical characteristics with increased surface area/volume ratio made them potential vectors to perform gene therapy. Various forms of nanoparticles are continued to be synthesised, and this review discusses the immediate barriers that nanoparticles have to encounter both during systemic movement in the body and intracellular trafficking to deliver the genes at the site of action.
Collapse
Affiliation(s)
- Susan Muthe Alex
- Facility for Nano/Microparticles Based Biomaterials for Advanced Drug Delivery Systems (FADDS) Division of Biosurface Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Poojappura, Thiruvananthapuram, Kerala, 695012, India
| | | |
Collapse
|
24
|
Adenoviral-mediated imaging of gene transfer using a somatostatin receptor-cytosine deaminase fusion protein. Cancer Gene Ther 2015; 22:215-21. [PMID: 25837665 PMCID: PMC4409539 DOI: 10.1038/cgt.2015.14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 01/13/2023]
Abstract
Suicide gene therapy is a process by which cells are administered a gene that encodes a protein capable of converting a nontoxic prodrug into an active toxin. Cytosine deaminase (CD) has been widely investigated as a means of suicide gene therapy due to the enzyme’s ability to convert the prodrug 5-fluorocytosine (5-FC) into the toxic compound 5-fluorouracil (5-FU). However, the extent of gene transfer is a limiting factor in predicting therapeutic outcome. The ability to monitor gene transfer, non-invasively, would strengthen the efficiency of therapy. In this regard, we have constructed and evaluated a replication-deficient adenovirus (Ad) containing the human somatostatin receptor subtype 2 (SSTR2) fused with a C-terminal yeast CD gene for the non-invasive monitoring of gene transfer and therapy. The resulting Ad (AdSSTR2-yCD) was evaluated in vitro in breast cancer cells to determine the function of the fusion protein. These studies demonstrated that the both the SSTR2 and yCD were functional in binding assays, conversion assays, and cytotoxicity assays. In vivo studies similarly demonstrated the functionality using conversion assays, biodistribution studies, and small animal positron-emission tomography (PET) imaging studies. In conclusion, the fusion protein has been validated as useful for the non-invasive imaging of yCD expression and will be evaluated in the future for monitoring yCD-based therapy.
Collapse
|
25
|
Penet MF, Chen Z, Li C, Winnard PT, Bhujwalla ZM. Prodrug enzymes and their applications in image-guided therapy of cancer: tracking prodrug enzymes to minimize collateral damage. Drug Deliv Transl Res 2015; 2:22-30. [PMID: 23646292 DOI: 10.1007/s13346-011-0052-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many cytotoxic therapies are available to kill cancer cells. Unfortunately, these also inflict significant damage on normal cells. Identifying highly effective cancer treatments that have minimal or no side effects continues to be a major challenge. One of the strategies to minimize damage to normal tissue is to deliver an activating enzyme that localizes only in the tumor and converts a nontoxic prodrug to a cytotoxic agent locally in the tumor. Such strategies have been previously tested but with limited success due in large part to the uncertainty in the delivery and distribution of the enzyme. Imaging the delivery of the enzyme to optimize timing of the prodrug administration to achieve image-guided prodrug therapy would be of immense benefit for this strategy. Here, we have reviewed advances in the incorporation of image guidance in the applications of prodrug enzymes in cancer treatment. These advances demonstrate the feasibility of using clinically translatable imaging in these prodrug enzyme strategies.
Collapse
Affiliation(s)
- Marie-France Penet
- JHU ICMIC Program, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
26
|
Verwilst P, Park S, Yoon B, Kim JS. Recent advances in Gd-chelate based bimodal optical/MRI contrast agents. Chem Soc Rev 2015; 44:1791-806. [DOI: 10.1039/c4cs00336e] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent developments in the field of bimodal MRI/optical contrast agents, based on Gd3+-chelates are presented.
Collapse
Affiliation(s)
- Peter Verwilst
- Department of Chemistry
- Korea University
- Seoul 136-701
- Korea
| | - Soyeon Park
- Department of Chemistry
- Korea University
- Seoul 136-701
- Korea
| | - Byungkwon Yoon
- Department of Chemistry
- Korea University
- Seoul 136-701
- Korea
| | - Jong Seung Kim
- Department of Chemistry
- Korea University
- Seoul 136-701
- Korea
| |
Collapse
|
27
|
Yang C, Guo W, An N, Cui L, Zhang T, Tong R, Chen Y, Lin H, Qu F. Enzyme-sensitive magnetic core–shell nanocomposites for triggered drug release. RSC Adv 2015. [DOI: 10.1039/c5ra15026d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Sodium hyaluronic acid cross-linked gel was employed to coat outside of Fe3O4@mSiO2 nanopaticles to prevent drug pervasion by a novel approach.
Collapse
Affiliation(s)
- Chunyu Yang
- College of Chemistry and Chemical Engineering
- Harbin Normal University
- Harbin 150025
- P. R. China
| | - Wei Guo
- Key Laboratory of Microsystems and Microstructures Manufacturing
- Ministry of Education
- Harbin Institute of Technology
- Harbin 150080
- P. R. China
| | - Na An
- College of Chemistry and Chemical Engineering
- Harbin Normal University
- Harbin 150025
- P. R. China
| | - Liru Cui
- College of Chemistry and Chemical Engineering
- Harbin Normal University
- Harbin 150025
- P. R. China
| | - Ting Zhang
- College of Chemistry and Chemical Engineering
- Harbin Normal University
- Harbin 150025
- P. R. China
| | - Ruihan Tong
- College of Chemistry and Chemical Engineering
- Harbin Normal University
- Harbin 150025
- P. R. China
| | - Yuhua Chen
- College of Chemistry and Chemical Engineering
- Harbin Normal University
- Harbin 150025
- P. R. China
| | - Huiming Lin
- College of Chemistry and Chemical Engineering
- Harbin Normal University
- Harbin 150025
- P. R. China
| | - Fengyu Qu
- College of Chemistry and Chemical Engineering
- Harbin Normal University
- Harbin 150025
- P. R. China
| |
Collapse
|
28
|
Wang J, Wang TT, Gao PF, Huang CZ. Biomolecules-conjugated nanomaterials for targeted cancer therapy. J Mater Chem B 2014; 2:8452-8465. [PMID: 32262204 DOI: 10.1039/c4tb01263a] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Biomolecules perform vital functions in biology. These functional biomolecules with diverse modifications hold great promise for further applications in bioanalysis and cancer therapy. However, these functional biomolecules face challenges, especially in the field of drug delivery for cancer therapy. For example, functional biomolecules are typically unstable when taken up by cells, as they are easily digested by enzymes. To address this obstacle, nanomaterials have been employed as drug carriers or vehicles, which are powerful nanoplatforms for imaging and cancer treatment. Multifunctionality of these nanoplatforms offers great advantages over conventional reagents, including targeting to a diseased site to minimize systemic toxicity, and the ability to solubilize hydrophobic or labile drugs to improved pharmacokinetics. In this review, we summarize typical functional biomolecule-conjugated nanomaterials for targeting drug delivery. Under the appropriate conditions, targeted drug delivery can be achieved from a high density of biomolecules that are bound to the surface of nanomaterials, resulting in a high affinity for the targets. The high density of biomolecules then leads to a high local concentration, being able to prevent degradation by enzymes. Furthermore, biomolecule-nanomaterial conjugates have been identified to enter cells more easily than free biomolecules, and controllable drug release can then be obtained by a response to a stimulus, such as redox, pH, light, thermal, enzyme-trigged strategies. Now and in the future, with the development of artificial biomolecules as well as nanomaterials, targeted drug delivery based on elegant biomolecule-nanomaterial conjugation approaches is expected to achieve great versatility, additional functions, and further advances.
Collapse
Affiliation(s)
- Jian Wang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China.
| | | | | | | |
Collapse
|
29
|
Ku SH, Kim K, Choi K, Kim SH, Kwon IC. Tumor-targeting multifunctional nanoparticles for siRNA delivery: recent advances in cancer therapy. Adv Healthc Mater 2014; 3:1182-93. [PMID: 24577795 DOI: 10.1002/adhm.201300607] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/20/2014] [Indexed: 11/06/2022]
Abstract
RNA interference (RNAi) is a naturally occurring regulatory process that controls posttranscriptional gene expression. Small interfering RNA (siRNA), a common form of RNAi-based therapeutics, offers new opportunities for cancer therapy via silencing specific genes, which are associated to cancer progress. However, clinical applications of RNAi-based therapy are still limited due to the easy degradation of siRNA during body circulation and the difficulty in the delivery of siRNA to desired tissues and cells. Thus, there have been many efforts to develop efficient siRNA delivery systems, which protect siRNA from serum nucleases and deliver siRNA to the intracellular region of target cells. Here, the recent advances in siRNA nanocarriers, which possess tumor-targeting ability are reviewed; various nanoparticle systems and their antitumor effects are summarized. The development of multifunctional nanocarriers for theranostics or combinatorial therapy is also discussed.
Collapse
Affiliation(s)
- Sook Hee Ku
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
| | - Kuiwon Choi
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology; Korea University; Seoul 136-701 Republic of Korea
| |
Collapse
|
30
|
Nayak TR, Krasteva LK, Cai W. Multimodality imaging of RNA interference. Curr Med Chem 2014; 20:3664-75. [PMID: 23745567 DOI: 10.2174/0929867311320290012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 03/13/2013] [Accepted: 03/13/2013] [Indexed: 12/16/2022]
Abstract
The discovery of small interfering RNAs (siRNAs) and their potential to knock down virtually any gene of interest has ushered in a new era of RNA interference (RNAi). Clinical use of RNAi faces severe limitations due to inefficiency delivery of siRNA or short hairpin RNA (shRNA). Many molecular imaging techniques have been adopted in RNAi-related research for evaluation of siRNA/shRNA delivery, biodistribution, pharmacokinetics, and the therapeutic effect. In this review article, we summarize the current status of in vivo imaging of RNAi. The molecular imaging techniques that have been employed include bioluminescence/fluorescence imaging, magnetic resonance imaging/ spectroscopy, positron emission tomography, single-photon emission computed tomography, and various combinations of these techniques. Further development of non-invasive imaging strategies for RNAi, not only focusing on the delivery of siRNA/shRNA but also the therapeutic efficacy, is critical for future clinical translation. Rigorous validation will be needed to confirm that biodistribution of the carrier is correlated with that of siRNA/shRNA, since imaging only detects the label (e.g. radioisotopes) but not the gene or carrier themselves. It is also essential to develop multimodality imaging approaches for realizing the full potential of therapeutic RNAi, as no single imaging modality may be sufficient to simultaneously monitor both the gene delivery and silencing effect of RNAi.
Collapse
Affiliation(s)
- T R Nayak
- Department of Radiology, University of Wisconsin - Madison, Madison, WI 53705-2275, USA
| | | | | |
Collapse
|
31
|
Bokacheva L, Ackerstaff E, LeKaye HC, Zakian K, Koutcher JA. High-field small animal magnetic resonance oncology studies. Phys Med Biol 2013; 59:R65-R127. [PMID: 24374985 DOI: 10.1088/0031-9155/59/2/r65] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This review focuses on the applications of high magnetic field magnetic resonance imaging (MRI) and spectroscopy (MRS) to cancer studies in small animals. High-field MRI can provide information about tumor physiology, the microenvironment, metabolism, vascularity and cellularity. Such studies are invaluable for understanding tumor growth and proliferation, response to treatment and drug development. The MR techniques reviewed here include (1)H, (31)P, chemical exchange saturation transfer imaging and hyperpolarized (13)C MRS as well as diffusion-weighted, blood oxygen level dependent contrast imaging and dynamic contrast-enhanced MRI. These methods have been proven effective in animal studies and are highly relevant to human clinical studies.
Collapse
Affiliation(s)
- Louisa Bokacheva
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, 415 East 68 Street, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
32
|
Penet MF, Artemov D, Farahani K, Bhujwalla ZM. MR - eyes for cancer: looking within an impenetrable disease. NMR IN BIOMEDICINE 2013; 26:745-55. [PMID: 23784955 PMCID: PMC3690531 DOI: 10.1002/nbm.2980] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 03/28/2013] [Accepted: 05/09/2013] [Indexed: 05/16/2023]
Abstract
Probe development is a critical component in cancer imaging, and novel probes are making major inroads in several aspects of cancer detection and image-guided treatments. Intrinsic MR probes such as signals from metabolites and their chemical shifts have been used for more than a decade to understand cancer physiology and metabolism. Through the integration of technology, molecular biology, and chemistry, the last few years have witnessed an explosion of extrinsic probes for molecular and functional imaging of cancer that, together with techniques such as CEST and hyperpolarization, have significantly expanded the repertoire of MR techniques in basic and translational investigations of many different aspects of cancer. Furthermore, incorporation of MR probes into multifunctional nanoparticles and multimodality imaging platforms have opened new opportunities for MR in image-guided diagnosis and therapy of cancer. Here we have provided an overview of recent innovations that have occurred in the development of MRI probes for molecular and functional imaging of cancer. Although most of these novel probes are not clinically available, they offer significant promise for future translational applications. In this review, we have highlighted the areas of future development that are likely to have a profound impact on cancer detection and treatment.
Collapse
Affiliation(s)
- Marie-France Penet
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Dmitri Artemov
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Keyvan Farahani
- Image Guided Interventions Branch, Cancer Imaging Program, National Cancer Institute, Bethesda MD, USA
| | - Zaver M. Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Correspondence to: Zaver M. Bhujwalla, Ph.D., Department of Radiology, Johns Hopkins University School of Medicine, 208C Traylor Building, 720 Rutland Avenue, Baltimore, MD 21205, USA., Phone: 410-955-9698, Fax: 410-614-1948,
| |
Collapse
|
33
|
Kim HO, Kim E, An Y, Choi J, Jang E, Choi EB, Kukreja A, Kim MH, Kang B, Kim DJ, Suh JS, Huh YM, Haam S. A Biodegradable Polymersome Containing Bcl-xL siRNA and Doxorubicin as a Dual Delivery Vehicle for a Synergistic Anticancer Effect. Macromol Biosci 2013; 13:745-54. [DOI: 10.1002/mabi.201200448] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/21/2013] [Indexed: 01/01/2023]
|
34
|
Chen Z, Penet MF, Nimmagadda S, Li C, Banerjee SR, Winnard PT, Artemov D, Glunde K, Pomper MG, Bhujwalla ZM. PSMA-targeted theranostic nanoplex for prostate cancer therapy. ACS NANO 2012; 6:7752-7762. [PMID: 22866897 PMCID: PMC4066818 DOI: 10.1021/nn301725w] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Theranostic imaging, where diagnosis is combined with therapy, is particularly suitable for a disease that is as complex as cancer, especially now that genomic and proteomic profiling can provide an extensive "fingerprint" of each tumor. With such information, theranostic agents can be designed to personalize treatment and minimize damage to normal tissue. Here we have developed a nanoplex platform for theranostic imaging of prostate cancer (PCa). In these proof-of-principle studies, a therapeutic nanoplex containing multimodal imaging reporters was targeted to prostate-specific membrane antigen (PSMA), which is expressed on the cell surface of castrate-resistant PCa. The nanoplex was designed to deliver small interfering RNA (siRNA) along with a prodrug enzyme to PSMA-expressing tumors. Each component of the nanoplex was carefully selected to evaluate its diagnostic aspect of PSMA imaging and its therapeutic aspects of siRNA-mediated down-regulation of a target gene and the conversion of a prodrug to cytotoxic drug, using noninvasive multimodality imaging. Studies performed using two variants of human PC3-PCa cells and tumors, one with high PSMA expression level and another with negligible expression levels, demonstrated PSMA-specific uptake. In addition, down-regulation of the selected siRNA target, choline kinase (Chk), and the conversion of the nontoxic prodrug 5-fluorocytosine (5-FC) to cytotoxic 5-fluorouracil (5-FU) were also demonstrated with noninvasive imaging. The nanoplex was well-tolerated and did not induce liver or kidney toxicity or a significant immune response. The nanoplex platform described can be easily modified and applied to different cancers, receptors, and pathways to achieve theranostic imaging, as a single agent or in combination with other treatment modalities.
Collapse
Affiliation(s)
- Zhihang Chen
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
| | - Marie-France Penet
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
| | - Sridhar Nimmagadda
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Cong Li
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
| | - Sangeeta R Banerjee
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
| | - Paul T Winnard
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
| | - Dmitri Artemov
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Kristine Glunde
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Martin G Pomper
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Zaver M Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
35
|
Shim MS, Kwon YJ. Stimuli-responsive polymers and nanomaterials for gene delivery and imaging applications. Adv Drug Deliv Rev 2012; 64:1046-59. [PMID: 22329941 DOI: 10.1016/j.addr.2012.01.018] [Citation(s) in RCA: 283] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 01/30/2012] [Accepted: 01/30/2012] [Indexed: 12/11/2022]
Abstract
Multiple extra- and intracellular obstacles, including low stability in blood, poor cellular uptake, and inefficient endosomal escape and disassembly in the cytoplasm, have to be overcome in order to deliver nucleic acids for gene therapy. This review introduces the recent advances in tackling the key challenges in achieving efficient, targeted, and safe nonviral gene delivery using various nucleic acid-containing nanomaterials that are designed to respond to various extra- and intracellular biological stimuli (e.g., pH, redox potential, and enzyme) as well as external artificial triggers (e.g., light and ultrasound). Gene delivery in combination with molecular imaging and targeting enables diagnostic assessment, treatment monitoring and quantification of efficiency, and confirmation of cure, thus fulfilling the great promise of efficient and personalized medicine. Nanomaterials platform for combined imaging and gene therapy, nanotheragnostics, using stimuli-responsive materials is also highlighted in this review. It is clear that developing novel multifunctional nonviral vectors, which transform their physico-chemical properties in response to various stimuli in a timely and spatially controlled manner, is highly desired to translate the promise of gene therapy for the clinical success.
Collapse
|
36
|
Abstract
Gene therapy has long been regarded a promising treatment for many diseases, whether acquired (such as AIDS or cancer) or inherited through a genetic disorder. A drug based on a nucleic acid, however, must be delivered to the interior of the target cell while surviving an array of biological defenses honed by evolution. Successful gene therapy is thus dependent on the development of an efficient delivery vector. Researchers have pursued two major vehicles for gene delivery: viral and nonviral (synthetic) vectors. Although viral vectors currently offer greater efficiency, nonviral vectors, which are typically based on cationic lipids or polymers, are preferred because of safety concerns with viral vectors. So far, nonviral vectors can readily transfect cells in culture, but efficient nanomedicines remain far removed from the clinic. Overcoming the obstacles associated with nonviral vectors to improve the delivery efficiency and therapeutic effect of nucleic acids is thus an active area of current research. The difficulties are manifold, including the strong interaction of cationic delivery vehicles with blood components, uptake by the reticuloendothelial system (RES), toxicity, and managing the targeting ability of the carriers with respect to the cells of interest. Modifying the surface with poly(ethylene glycol), that is, PEGylation, is the predominant method used to reduce the binding of plasma proteins to nonviral vectors and minimize clearance by the RES after intravenous administration. Nanoparticles that are not rapidly cleared from the circulation accumulate in the tumors because of the enhanced permeability and retention effect, and the targeting ligands attached to the distal end of the PEGylated components allow binding to the receptors on the target cell surface. Neutral and anionic liposomes have been also developed for systemic delivery of nucleic acids in experimental animal models. Other approaches include (i) designing and synthesizing novel cationic lipids and polymers, (ii) chemically coupling the nucleic acid to peptides, targeting ligands, polymers, or environmentally sensitive moieties, and (iii) utilizing inorganic nanoparticles in nucleic acid delivery. Recently, the different classes of nonviral vectors appear to be converging, and the ability to combine features of different classes of nonviral vectors in a single strategy has emerged. With the strengths of several approaches working in concert, more hurdles associated with efficient nucleic acid delivery might therefore be overcome. In this Account, we focus on these novel nonviral vectors, which are classified as multifunctional hybrid nucleic acid vectors, novel membrane/core nanoparticles for nucleic acid delivery, and ultrasound-responsive nucleic acid vectors. We highlight systemic delivery studies and consider the future prospects for nucleic acid delivery. A better understanding of the fate of the nanoparticles inside the cell and of the interactions between the parts of hybrid particles should lead to a delivery system suitable for clinical use. We also underscore the value of sustained release of a nucleic acid in this endeavor; making vectors targeted to cells with sustained release in vivo should provide an interesting research challenge.
Collapse
Affiliation(s)
- Xia Guo
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, P. R. China
| | - Leaf Huang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
37
|
Suicide gene therapy in cancer: where do we stand now? Cancer Lett 2012; 324:160-70. [PMID: 22634584 DOI: 10.1016/j.canlet.2012.05.023] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 04/11/2012] [Accepted: 05/21/2012] [Indexed: 12/21/2022]
Abstract
Suicide gene therapy is based on the introduction into tumor cells of a viral or a bacterial gene, which allows the conversion of a non-toxic compound into a lethal drug. Although suicide gene therapy has been successfully used in a large number of in vitro and in vivo studies, its application to cancer patients has not reached the desirable clinical significance. However, recent reports on pre-clinical cancer models demonstrate the huge potential of this strategy when used in combination with new therapeutic approaches. In this review, we summarize the different suicide gene systems and gene delivery vectors addressed to cancer, with particular emphasis on recently developed systems and associated bystander effects. In addition, we review the different strategies that have been used in combination with suicide gene therapy and provide some insights into the future directions of this approach, particularly towards cancer stem cell eradication.
Collapse
|
38
|
Ifediba MA, Moore A. In vivo imaging of the systemic delivery of small interfering RNA. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2012; 4:428-37. [PMID: 22228711 DOI: 10.1002/wnan.1158] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Short interfering RNAs (siRNAs) have emerged as a potent new class of therapeutics, which regulate gene expression through sequence-specific inhibition of mRNA translation. Human trials of siRNAs have highlighted the need for robust delivery and detection techniques that will enable the application of these therapeutics to increasingly complex disease and organ systems. Efforts to monitor the in vivo trafficking and efficacy of siRNAs have routinely involved bioluminescence imaging of naked siRNA molecules. More recently, siRNAs have been incorporated into a variety of molecular imaging probes to promote their detection with clinically relevant imaging modalities. Lipid-, polymer-, and nanoparticle-based siRNA delivery vehicles have proven effective in improving the stability, bioavailability, and target specificity of siRNAs following systemic administration in vivo. Additionally, these methods provide a platform to modify siRNAs with a variety of contrast agents and have enabled nuclear and magnetic resonance imaging of siRNA delivery in preclinical studies. These image-guided delivery approaches represent a crucial step in the transition of siRNA therapeutics to the clinic.
Collapse
Affiliation(s)
- Marytheresa A Ifediba
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | | |
Collapse
|
39
|
Abstract
RNA interference (RNAi) has been extensively employed for in vivo research since its use was first demonstrated in mammalian cells 10 years ago. Design rules have improved, and it is now routinely possible to obtain reagents that suppress expression of any gene desired. At the same time, increased understanding of the molecular basis of unwanted side effects has led to the development of chemical modification strategies that mitigate these concerns. Delivery remains the single greatest hurdle to widespread adoption of in vivo RNAi methods. However, exciting advances have been made and new delivery systems under development may help to overcome these barriers. This review discusses advances in RNAi biochemistry and biology that impact in vivo use and provides an overview of select publications that demonstrate interesting applications of these principles. Emphasis is placed on work with synthetic, small interfering RNAs (siRNAs) published since the first installment of this review which appeared in 2006.
Collapse
|
40
|
Laurent S, Mahmoudi M. Superparamagnetic iron oxide nanoparticles: promises for diagnosis and treatment of cancer. INTERNATIONAL JOURNAL OF MOLECULAR EPIDEMIOLOGY AND GENETICS 2011; 2:367-390. [PMID: 22199999 PMCID: PMC3243452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 11/22/2011] [Indexed: 05/31/2023]
Abstract
During the last decade, significant scientific research efforts have led to a significant growth in understanding of cancer at the genetic, molecular, and cellular levels providing great opportunities for diagnosis and treatment of cancer diseases. The hopes for fast cancer diagnosis and treatment were significantly increased by the entrance of nanoparticles to the medical sciences. Nanoparticles are attractive due to their unique opportunities together with negligible side effects not only in cancer therapy but also in the treatment of other ailments. Among all types of nanoparticles, surface-engineered superparamagnetic iron oxide nanoparticles (SPIONs) have been attracted a great attention for cancer therapy applications. This review covers the recent advances in the development of SPIONs together with their opportunities and challenges, as theranosis agents, in cancer treatment.
Collapse
Affiliation(s)
- Sophie Laurent
- Department of General, Organic, and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau19, B-7000 Mons, Belgium
| | - Morteza Mahmoudi
- National Cell Bank, Pasteur Institute of IranTehran, Iran
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical SciencesTehran, Iran
| |
Collapse
|
41
|
Abstract
Abnormal choline metabolism is emerging as a metabolic hallmark that is associated with oncogenesis and tumour progression. Following transformation, the modulation of enzymes that control anabolic and catabolic pathways causes increased levels of choline-containing precursors and breakdown products of membrane phospholipids. These increased levels are associated with proliferation, and recent studies emphasize the complex reciprocal interactions between oncogenic signalling and choline metabolism. Because choline-containing compounds are detected by non-invasive magnetic resonance spectroscopy (MRS), increased levels of these compounds provide a non-invasive biomarker of transformation, staging and response to therapy. Furthermore, enzymes of choline metabolism, such as choline kinase, present novel targets for image-guided cancer therapy.
Collapse
Affiliation(s)
- Kristine Glunde
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, The Russell H. Morgan Department of Radiology and Radiological Science, 720 Rutland Avenue, 212 Traylor Building, Baltimore, Maryland 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland 21231, USA
| | - Zaver M. Bhujwalla
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, The Russell H. Morgan Department of Radiology and Radiological Science, 720 Rutland Avenue, 212 Traylor Building, Baltimore, Maryland 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland 21231, USA
| | - Sabrina M. Ronen
- Department of Radiology, University of California San Francisco School of Medicine, UCSF Mission Bay Campus, Byers Hall, San Francisco, California CA94158-2330, USA
| |
Collapse
|
42
|
Cationic drug-derived nanoparticles for multifunctional delivery of anticancer siRNA. Biomaterials 2011; 32:9785-95. [PMID: 21937102 DOI: 10.1016/j.biomaterials.2011.09.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 09/07/2011] [Indexed: 01/27/2023]
Abstract
Combined treatment of anticancer drugs and small interfering RNAs (siRNAs) have emerged as a new modality of anticancer therapy. Here, we describe a co-delivery system of anticancer drugs and siRNA in which anticancer drug-derived lipids form cationic nanoparticles for siRNA complexation. The anticancer drug mitoxantrone (MTO) was conjugated to palmitoleic acid, generating two types of palmitoleyl MTO (Pal-MTO) lipids: monopalmitoleyl MTO (mono-Pal-MTO) and dipalmitoleyl MTO (di-Pal-MTO). Among various lipid compositions of MTO, nanoparticles containing mono-Pal-MTO and di-Pal-MTO at a molar ratio of 1:1 (md11-Pal-MTO nanoparticles) showed the most efficient cellular delivery of siRNA, higher than that of Lipofectamine 2000. Delivery of red fluorescence protein-specific siRNA into B16F10-RFP cells using md11-Pal-MTO nanoparticles reduced the expression of RFP at both mRNA and protein levels, demonstrating silencing of the siRNA target gene. Moreover, delivery of Mcl-1-specific anticancer siRNA (siMcl-1) using md11-Pal-MTO enhanced antitumor activity in vitro, reducing tumor cell viability by 81% compared to a reduction of 68% following Lipofectamine 2000-mediated transfection of siMcl-1. Intratumoral administration of siMcl-1 using md11-Pal-MTO nanoparticles significantly inhibited tumor growth, reducing tumor size by 83% compared to untreated controls. Our results suggest the potential of md11-Pal-MTO multifunctional nanoparticles for co-delivery of anticancer siRNAs for effective combination therapy.
Collapse
|
43
|
Stasinopoulos I, Penet MF, Krishnamachary B, Bhujwalla ZM. Molecular and functional imaging of invasion and metastasis: windows into the metastatic cascade. Cancer Biomark 2011; 7:173-88. [PMID: 21576811 DOI: 10.3233/cbm-2010-0188] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The ability of cancer cells to invade, metastasize, and form distant colonies, is one of the key characteristics that confers lethality to cancer. Metastatic cancer cells typically become refractory to treatment. The metastatic cascade is a multi-step process that is governed by events within the cancer cell, the tumor microenvironment, and the distant environments that are invaded and colonized by the cancer cells. Noninvasive imaging techniques are facilitating a close examination of the stepwise journey of the cancer cell from the primary tumor to the distant metastatic site. Here we have discussed the metastatic process, and how molecular and functional imaging of cancer are providing new insights into the metastatic cascade that can be exploited for treatment of metastatic disease.
Collapse
Affiliation(s)
- Ioannis Stasinopoulos
- JHU ICMIC Program, The Russell H. Morgan Department of Radiology and Radiological Science, USA
| | | | | | | |
Collapse
|
44
|
|