1
|
Pan Y, Tang W, Fan W, Zhang J, Chen X. Development of nanotechnology-mediated precision radiotherapy for anti-metastasis and radioprotection. Chem Soc Rev 2022; 51:9759-9830. [DOI: 10.1039/d1cs01145f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radiotherapy (RT), including external beam RT and internal radiation therapy, uses high-energy ionizing radiation to kill tumor cells.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Wei Tang
- Departments of Pharmacy and Diagnostic Radiology, Nanomedicine Translational Research Program, Faculty of Science and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
2
|
Lo WL, Lo SW, Chen SJ, Chen MW, Huang YR, Chen LC, Chang CH, Li MH. Molecular Imaging and Preclinical Studies of Radiolabeled Long-Term RGD Peptides in U-87 MG Tumor-Bearing Mice. Int J Mol Sci 2021; 22:ijms22115459. [PMID: 34064291 PMCID: PMC8196871 DOI: 10.3390/ijms22115459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/10/2021] [Accepted: 05/20/2021] [Indexed: 11/18/2022] Open
Abstract
The Arg–Gly–Asp (RGD) peptide shows a high affinity for αvβ3 integrin, which is overexpressed in new tumor blood vessels and many types of tumor cells. The radiolabeled RGD peptide has been studied for cancer imaging and radionuclide therapy. We have developed a long-term tumor-targeting peptide DOTA-EB-cRGDfK, which combines a DOTA chelator, a truncated Evans blue dye (EB), a modified linker, and cRGDfK peptide. The aim of this study was to evaluate the potential of indium-111(111In) radiolabeled DOTA-EB-cRGDfK in αvβ3 integrin-expressing tumors. The human glioblastoma cell line U-87 MG was used to determine the in vitro binding affinity of the radiolabeled peptide. The in vivo distribution of radiolabeled peptides in U-87 MG xenografts was investigated by biodistribution, nanoSPECT/CT, pharmacokinetic and excretion studies. The in vitro competition assay showed that 111In-DOTA-EB-cRGDfK had a significant binding affinity to U-87 MG cancer cells (IC50 = 71.7 nM). NanoSPECT/CT imaging showed 111In-DOTA-EB-cRGDfK has higher tumor uptake than control peptides (111In-DOTA-cRGDfK and 111In-DOTA-EB), and there is still a clear signal until 72 h after injection. The biodistribution results showed significant tumor accumulation (27.1 ± 2.7% ID/g) and the tumor to non-tumor ratio was 22.85 at 24 h after injection. In addition, the pharmacokinetics results indicated that the 111In-DOTA-EB-cRGDfK peptide has a long-term half-life (T1/2λz = 77.3 h) and that the calculated absorbed dose was safe for humans. We demonstrated that radiolabeled DOTA-EB-cRGDfK may be a promising agent for glioblastoma tumor imaging and has the potential as a theranostic radiopharmaceutical.
Collapse
Affiliation(s)
- Wei-Lin Lo
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Shih-Wei Lo
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Su-Jung Chen
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Ming-Wei Chen
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Yuan-Ruei Huang
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Liang-Cheng Chen
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Chih-Hsien Chang
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Correspondence: (C.-H.C.); (M.-H.L.)
| | - Ming-Hsin Li
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
- Correspondence: (C.-H.C.); (M.-H.L.)
| |
Collapse
|
3
|
Translating Research for the Radiotheranostics of Nanotargeted 188Re-Liposome. Int J Mol Sci 2021; 22:ijms22083868. [PMID: 33918011 PMCID: PMC8068325 DOI: 10.3390/ijms22083868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Nanoliposomes are one of the leading potential nano drug delivery systems capable of targeting chemotherapeutics to tumor sites because of their passive nano-targeting capability through the enhanced permeability and retention (EPR) effect for cancer patients. Recent advances in nano-delivery systems have inspired the development of a wide range of nanotargeted materials and strategies for applications in preclinical and clinical usage in the cancer field. Nanotargeted 188Re-liposome is a unique internal passive radiotheranostic agent for nuclear imaging and radiotherapeutic applications in various types of cancer. This article reviews and summarizes our multi-institute, multidiscipline, and multi-functional studied results and achievements in the research and development of nanotargeted 188Re-liposome from preclinical cells and animal models to translational clinical investigations, including radionuclide nanoliposome formulation, targeted nuclear imaging, biodistribution, pharmacokinetics, radiation dosimetry, radiation tumor killing effects in animal models, nanotargeted radionuclide and radio/chemo-combination therapeutic effects, and acute toxicity in various tumor animal models. The systemic preclinical and clinical studied results suggest 188Re-liposome is feasible and promising for in vivo passive nanotargeted radionuclide theranostics in future cancer care applications.
Collapse
|
4
|
Tsang YW, Chi KH, Huang CC, Chi MS, Chiang HC, Yang KL, Li WT, Wang YS. Modulated electro-hyperthermia-enhanced liposomal drug uptake by cancer cells. Int J Nanomedicine 2019; 14:1269-1279. [PMID: 30863059 PMCID: PMC6391149 DOI: 10.2147/ijn.s188791] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Modulated electro-hyperthermia (mEHT) stands to be a significant technological advancement in the hyperthermia field, utilizing autofocusing electromagnetic power on the cell membrane to create massive apoptosis. Since mEHT possesses the unique ability to excite cell membranes, we hypothesized that mEHT could enhance the uptake of liposomal drugs by enhancing phagocytic activity. Materials and methods Water bath control and mEHT were used to compare the enhancement of liposome-encapsulated doxorubicin (Lipodox®) uptake by cancer cells. Cancer cells were made visible by doxorubicin fluorescence to investigate drug uptake. Viable cell yield was determined via the Trypan Blue exclusion method. Various substrates were used to investigate the mechanism of drug-uptake enhancement. The murine colon carcinoma model, CT26, was used to confirm the tissue infiltration of Lipodox® and its therapeutic effect. Results mEHT treatment showed a significant enhancement of Lipodox® uptake of doxorubicin fluorescence compared with 37°C or 42°C water bath treatment. Tumor tissue sections also confirmed that mEHT treatment achieved the highest doxorubicin concentration in vivo (1.44±0.32 µg/g in mEHT group and 0.79±0.32 µg/g in 42°C water bath). Wortmannin was used to inhibit the macropinocytosis effect and 70 kDa dextran-FITC served as uptake substance. The uptake of dextran-FITC by cancer cells significantly increased after mEHT treatment whereas such enhancement was significantly inhibited by wortmannin. Conclusion The result showed mEHT-induced particle-uptake through macropinocytosis. mEHT-enhanced uptake of Lipodox® may amplify the therapeutic effect of liposomal drugs. This novel finding warrants further clinical investigation.
Collapse
Affiliation(s)
- Yuk-Wah Tsang
- Department of Radiation Oncology, Chiayi Christian Hospital, Chiayi, Taiwan.,Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan, Taiwan,
| | - Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, .,Institute of Radiation Science and School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Chung Huang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, .,Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mau-Shin Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, .,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan,
| | - Hsin-Chien Chiang
- Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Kai-Lin Yang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, .,Institute of Radiation Science and School of Medicine, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wen-Tyng Li
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan, Taiwan,
| | - Yu-Shan Wang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, .,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan,
| |
Collapse
|
5
|
Chang YJ, Ho CL, Cheng KH, Kuo WI, Lee WC, Lan KL, Chang CH. Biodistribution, pharmacokinetics and radioimmunotherapy of 188Re-cetuximab in NCI-H292 human lung tumor-bearing nude mice. Invest New Drugs 2019; 37:961-972. [PMID: 30612308 DOI: 10.1007/s10637-018-00718-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/25/2018] [Indexed: 02/07/2023]
Abstract
Background Cetuximab is a fully humanized IgG1 subclass monoclonal that binds specifically to the human epidermal growth factor receptor (EGFR). Although EGFR is expressed in normal cells, the overexpression of EGFR is detected in many human cancers, such as colon, rectum and lung tumors. In this study, cetuximab with a combination of radiotherapy nuclear 188Re achieved better therapeutic effect on lung cancer. Methods188Re-cetuximab administered by the i.v. route in human NCI-H292 lung tumor-bearing mice was investigated. NanoSPECT/CT images were taken to evaluate the distribution and tumor targeting of 188Re-cetuximab in mice. The anti-tumor effect of 188Re-cetuximab was assessed by the tumor growth inhibition, survival ratio. Results For nanoSPECT/CT imaging, a significant uptake in tumor was observed at 24 and 48 h following the injection of 188Re-cetuximab. The anti-tumor effect of 188Re-cetuximab was assessed by tumor growth inhibition and the survival ratio. The tumor-bearing mice treated with 188Re-cetuximab showed a better mean tumor growth inhibition rate (MGI = 0.049) and longer median survival time and lifespan (62.50 d; 70.07%) than those treated with 188Re-perrhenate and cetuximab only by single injection. A synergistic effect of tumor growth inhibition was observed with the combination index exceeding one for 188Re-cetuximab (CI = 6.135 and 9.276). Conclusion The tumor targeting and localization of 188Re-cetuximab were confirmed in this study. Synergistic therapeutic efficacy was demonstrated for the radioimmunotherapy of 188Re-cetuximab. The results of this study reveal the potential advantage and benefit obtained from 188Re-cetuximab for diagnosis and therapy of oncology applications in the future.
Collapse
Affiliation(s)
- Ya-Jen Chang
- Institute of Nuclear Energy Research, 1000 Wenhua Rd, Longtan District, Taoyuan City, Taiwan
| | - Chung-Li Ho
- Institute of Nuclear Energy Research, 1000 Wenhua Rd, Longtan District, Taoyuan City, Taiwan
| | - Kai-Hung Cheng
- Institute of Nuclear Energy Research, 1000 Wenhua Rd, Longtan District, Taoyuan City, Taiwan
| | - Wan-I Kuo
- Institute of Nuclear Energy Research, 1000 Wenhua Rd, Longtan District, Taoyuan City, Taiwan
| | - Wan-Chi Lee
- Institute of Nuclear Energy Research, 1000 Wenhua Rd, Longtan District, Taoyuan City, Taiwan
| | - Keng-Li Lan
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Hsien Chang
- Institute of Nuclear Energy Research, 1000 Wenhua Rd, Longtan District, Taoyuan City, Taiwan.
| |
Collapse
|
6
|
Song G, Cheng L, Chao Y, Yang K, Liu Z. Emerging Nanotechnology and Advanced Materials for Cancer Radiation Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1700996. [PMID: 28643452 DOI: 10.1002/adma.201700996] [Citation(s) in RCA: 449] [Impact Index Per Article: 64.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 04/11/2017] [Indexed: 05/22/2023]
Abstract
Radiation therapy (RT) including external beam radiotherapy (EBRT) and internal radioisotope therapy (RIT) has been widely used for clinical cancer treatment. However, owing to the low radiation absorption of tumors, high doses of ionizing radiations are often needed during RT, leading to severe damages to normal tissues adjacent to tumors. Meanwhile, the RT efficacies are limited by different mechanisms, among which the tumor hypoxia-associated radiation resistance is a well-known one, as there exists hypoxia inside most solid tumors while oxygen is essential to enhance radiation-induced DNA damages. With the development in nanotechnology, there have been great interests in using nanomedicine strategies to enhance radiation responses of tumors. Nanomaterials containing high-Z elements to absorb radiation rays (e.g. X-ray) can act as radio-sensitizers to deposit radiation energy within tumors and promote treatment efficacy. Nanoscale carriers are able to deliver therapeutic radioisotopes into tumors for internal RIT, or chemotherapeutic drugs for synergistically combined chemo-radiotherapy. As uncovered in recent studies, the tumor microenvironment could be modulated by various nanomedicine approaches to overcome hypoxia-associated radiation resistance. Herein, the authors will summarize the applications of nanomedicine for RT cancer treatment, and pay particular attention to the latest development of 'advanced materials' for enhanced cancer RT.
Collapse
Affiliation(s)
- Guosheng Song
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California, 94305-5484, USA
| | - Liang Cheng
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yu Chao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kai Yang
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
7
|
Zhou M, Chen Y, Adachi M, Wen X, Erwin B, Mawlawi O, Lai SY, Li C. Single agent nanoparticle for radiotherapy and radio-photothermal therapy in anaplastic thyroid cancer. Biomaterials 2015; 57:41-9. [PMID: 25913249 DOI: 10.1016/j.biomaterials.2015.04.013] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 04/01/2015] [Accepted: 04/03/2015] [Indexed: 02/01/2023]
Abstract
Anaplastic thyroid carcinoma (ATC) is one of the most aggressive human malignancies. The aggressive behavior of ATC and its resistance to traditional treatment limit the efficacy of radiotherapy, chemotherapy, and surgery. The purpose of this study is aimed at enhancing the therapeutic efficacy of radiotherapy (RT) combined with photothermal therapy (PTT) in murine orthotopic model of ATC, based on our developed single radioactive copper sulfide (CuS) nanoparticle platform. We prepare a new dual-modality therapy for ATC consisting of a single-compartment nanoplatform, polyethylene glycol-coated [(64)Cu]CuS NPs, in which the radiotherapeutic property of (64)Cu is combined with the plasmonic properties of CuS NPs. Mice with Hth83 ATC were treated with PEG-[(64)Cu]CuS NPs and/or near infrared laser. Antitumor effects were assessed by tumor growth and animal survival. We found that in mice bearing orthotopic human Hth83 ATC tumors, micro-PET/CT imaging and biodistribution studies showed that about 50% of the injected dose of PEG-[(64)Cu]CuS NPs was retained in tumor 48 h after intratumoral injection. Human absorbed doses were calculated from biodistribution data. In antitumor experiments, tumor growth was delayed by PEG-[(64)Cu]CuS NP-mediated RT, PTT, and combined RT/PTT, with combined RT/PTT being most effective. In addition, combined RT/PTT significantly prolonged the survival of Hth83 tumor-bearing mice compared to no treatment, laser treatment alone, or NP treatment alone without producing acute toxic effects. These findings indicate that this single-compartment multifunctional NPs platform merits further development as a novel therapeutic agent for ATC.
Collapse
Affiliation(s)
- Min Zhou
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yunyun Chen
- Department of Head and Neck Surgery, and Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Makoto Adachi
- Department of Head and Neck Surgery, and Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaoxia Wen
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bill Erwin
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Osama Mawlawi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Stephen Y Lai
- Department of Head and Neck Surgery, and Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Gaddy DF, Lee H, Zheng J, Jaffray DA, Wickham TJ, Hendriks BS. Whole-body organ-level and kidney micro-dosimetric evaluations of (64)Cu-loaded HER2/ErbB2-targeted liposomal doxorubicin ((64)Cu-MM-302) in rodents and primates. EJNMMI Res 2015; 5:24. [PMID: 25918676 PMCID: PMC4404468 DOI: 10.1186/s13550-015-0096-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/08/2015] [Indexed: 02/04/2023] Open
Abstract
Background Features of the tumor microenvironment influence the efficacy of cancer nanotherapeutics. The ability to directly radiolabel nanotherapeutics offers a valuable translational tool to obtain biodistribution and tumor deposition data, testing the hypothesis that the extent of delivery predicts therapeutic outcome. In support of a first in-human clinical trial with 64Cu-labeled HER2-targeted liposomal doxorubicin (64Cu-MM-302), a preclinical dosimetric analysis was performed. Methods Whole-body biodistribution and pharmacokinetic data were obtained in mice that received 64Cu-MM-302 and used to estimate absorbed radiation doses in normal human organs. PET/CT imaging revealed non-uniform distribution of 64Cu signal in mouse kidneys. Kidney micro-dosimetry analysis was performed in mice and squirrel monkeys, using a physiologically based pharmacokinetic model to estimate the full dynamics of the 64Cu signal in monkeys. Results Organ-level dosimetric analysis of mice receiving 64Cu-MM-302 indicated that the heart was the organ receiving the highest radiation absorbed dose, due to extended liposomal circulation. However, PET/CT imaging indicated that 64Cu-MM-302 administration resulted in heterogeneous exposure in the kidney, with a focus of 64Cu activity in the renal pelvis. This result was reproduced in primates. Kidney micro-dosimetry analysis illustrated that the renal pelvis was the maximum exposed tissue in mice and squirrel monkeys, due to the highly concentrated signal within the small renal pelvis surface area. Conclusions This study was used to select a starting clinical radiation dose of 64Cu-MM-302 for PET/CT in patients with advanced HER2-positive breast cancer. Organ-level dosimetry and kidney micro-dosimetry results predicted that a radiation dose of 400 MBq of 64Cu-MM-302 should be acceptable in patients. Electronic supplementary material The online version of this article (doi:10.1186/s13550-015-0096-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel F Gaddy
- Merrimack Pharmaceuticals, One Kendall Square, Suite B7201, Cambridge, MA 02139 USA
| | - Helen Lee
- Merrimack Pharmaceuticals, One Kendall Square, Suite B7201, Cambridge, MA 02139 USA
| | - Jinzi Zheng
- STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, 190 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| | - David A Jaffray
- STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, 190 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| | - Thomas J Wickham
- Merrimack Pharmaceuticals, One Kendall Square, Suite B7201, Cambridge, MA 02139 USA
| | - Bart S Hendriks
- Merrimack Pharmaceuticals, One Kendall Square, Suite B7201, Cambridge, MA 02139 USA
| |
Collapse
|
9
|
Huang FYJ, Lee TW, Chang CH, Chen LC, Hsu WH, Chang CW, Lo JM. Evaluation of 188Re-labeled PEGylated nanoliposome as a radionuclide therapeutic agent in an orthotopic glioma-bearing rat model. Int J Nanomedicine 2015; 10:463-73. [PMID: 25624760 PMCID: PMC4296959 DOI: 10.2147/ijn.s75955] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE In this study, the (188)Re-labeled PEGylated nanoliposome ((188)Re-liposome) was prepared and evaluated as a therapeutic agent for glioma. MATERIALS AND METHODS The reporter cell line, F98(luc) was prepared via Lentivector expression kit system and used to set up the orthotopic glioma-bearing rat model for non-invasive bioluminescent imaging. The maximum tolerated dose applicable in Fischer344 rats was explored via body weight monitoring of the rats after single intravenous injection of (188)Re-liposome with varying dosages before the treatment study. The OLINDA/EXM 1.1 software was utilized for estimating the radiation dosimetry. To assess the therapeutic efficacy, tumor-bearing rats were intravenously administered (188)Re-liposome or normal saline followed by monitoring of the tumor growth and animal survival time. In addition, the histopathological examinations of tumors were conducted on the (188)Re-liposome-treated rats. RESULTS By using bioluminescent imaging, the well-established reporter cell line (F98(luc)) showed a high relationship between cell number and its bioluminescent intensity (R(2)=0.99) in vitro; furthermore, it could also provide clear tumor imaging for monitoring tumor growth in vivo. The maximum tolerated dose of (188)Re-liposome in Fischer344 rats was estimated to be 333 MBq. According to the dosimetry results, higher equivalent doses were observed in spleen and kidneys while very less were in normal brain, red marrow, and thyroid. For therapeutic efficacy study, the progression of tumor growth in terms of tumor volume and/or tumor weight was significantly slower for the (188)Re-liposome-treated group than the control group (P<0.05). As a result, the lifespan of glioma-bearing rats treated with (188)Re-liposome was prolonged 10.67% compared to the control group. CONCLUSION The radiotherapeutic evaluation by dosimetry and survival studies have demonstrated that passive targeting (188)Re-liposome via systemic administration can significantly prolong the lifespan of orthotopic glioma-bearing rats while maintaining reasonable systemic radiation safety. Therefore, (188)Re-liposome could be a potential therapeutic agent for glioblastoma multiforme treatment.
Collapse
Affiliation(s)
- Feng-Yun J Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Te-Wei Lee
- Institute of Nuclear Energy Research, Longtan, Taiwan
| | | | | | - Wei-Hsin Hsu
- Institute of Nuclear Energy Research, Longtan, Taiwan
| | - Chien-Wen Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Jem-Mau Lo
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
10
|
Liu SY, Chang CH, Lee TW. Single dose acute toxicity testing for N,N-bis(2-mercaptoethyl)-N',N' diethylethylenediamine in beagles. Regul Toxicol Pharmacol 2014; 69:217-25. [PMID: 24721389 DOI: 10.1016/j.yrtph.2014.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 03/29/2014] [Accepted: 04/01/2014] [Indexed: 10/25/2022]
Abstract
N,N-Bis(2-mercaptoethyl)-N',N'-diethylenediamine (BMEDA) is used in the preparation of the (188)Re-BMEDA-liposome as a chelator to deliver rhenium 188 into liposomes. Although the safety of the (188)Re-BMEDA-liposome had been established, the use of BMEDA in preparing the liposome is of interest; however, an assessment of its safety is warranted. In this present work, we report on the acute toxicity study of BMEDA in beagles to identify doses causing no adverse effect and doses causing life-threatening toxicity. In a single dose 14-day systemic toxicity study conducted in beagles, BMEDA was without compound-related adverse effects at doses of up to 2mg/kg in a series of clinical observations and clinical pathology examinations. The results of these studies could aid in choosing doses for repeat-dose studies and in the selection of starting doses for Phase 1 human studies.
Collapse
Affiliation(s)
- Si-Yen Liu
- Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan, Taiwan
| | - Chih-Hsien Chang
- Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan, Taiwan
| | - Te-Wei Lee
- Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan, Taiwan.
| |
Collapse
|
11
|
Chang CH, Liu SY, Lee TW. Pharmacokinetics of BMEDA after intravenous administration in beagle dogs. Molecules 2014; 19:538-49. [PMID: 24394437 PMCID: PMC6271955 DOI: 10.3390/molecules19010538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/02/2013] [Accepted: 12/03/2013] [Indexed: 01/17/2023] Open
Abstract
The pharmacokinetics of N,N-bis(2-mercapatoethly)-N',N'-diethylenediamine (BMEDA), a molecule that can form a chelate with rhenium-188 (188Re) to produce the 188Re-BMEDA-liposomes, was studied. In this work, beagles received a single injection of BMEDA, at doses of 1, 2, or 5 mg/kg; the concentration of BMEDA in the beagles' plasma was then analyzed and determined by liquid chromatography-mass spectrometry/mass spectrometry. Based on the pharmacokinetic parameters of BMEDA, we found that male and female animals shared similar patterns indicating that the pharmacokinetics of BMEDA is independent of gender differences. In addition, the pharmacokinetics of BMEDA was seen to be non-linear because the increase of mean AUC0-t and AUC0-∞ values tend to be greater than dose proportional while the mean Vss and CL values of BMEDA appeared to be dose dependent. The information on the pharmacokinetics of BMEDA generated from this study will serve as a basis to design appropriate pharmacology and toxicology studies for future human use.
Collapse
Affiliation(s)
- Chih-Hsien Chang
- Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan.
| | - Si-Yen Liu
- Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan.
| | - Te-Wei Lee
- Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan.
| |
Collapse
|
12
|
Hsu CW, Chang YJ, Chang CH, Chen LC, Lan KL, Ting G, Lee TW. Comparative therapeutic efficacy of rhenium-188 radiolabeled-liposome and 5-fluorouracil in LS-174T human colon carcinoma solid tumor xenografts. Cancer Biother Radiopharm 2013; 27:481-9. [PMID: 23067100 DOI: 10.1089/cbr.2011.1158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Nanoliposomes are important carriers capable of packaging drugs for various delivery applications. Rhenium-188-radiolabeled liposome ((188)Re-liposome) has potential for radiotherapy and diagnostic imaging. To evaluate the targeting of (188)Re-liposome, biodistribution, microSPECT/CT, whole-body autoradiography (WBAR), and pharmacokinetics were performed in LS-174T human tumor-bearing mice. The comparative therapeutic efficacy of (188)Re-liposome and 5-fluorouracil (5-FU) was assessed according to inhibition of tumor growth and the survival ratio. The highest uptake of (188)Re-liposome in LS-174T tumor was found at 24 hours by biodistribution and microSPECT/CT imaging, showing a positive correlation for tumor targeting of (188)Re-liposome using the Pearson's correlation analysis (r=0.997). Pharmacokinetics of (188)Re-liposome showed the properties of high circulation time and high bioavailability (mean residence time [MRT]=18.8 hours, area under the curve [AUC]=1371%ID/g·h). For therapeutic efficacy, the tumor-bearing mice treated with (188)Re-liposome (80% maximum tolerated dose [MTD], 23.7 MBq) showed better tumor growth inhibition and longer survival time than those treated with 5-FU (80% MTD, 144 mg/kg). The median survival time for mice treated with (188)Re-liposome (58.5 days; p<0.05) was significantly better than those of 5-FU (48.25 days; p>0.05) and normal saline-treated mice (43.63 days). Dosimetry study revealed that the (188)Re-liposome did not lead to high absorbed doses in normal tissue, but did in small tumors. These results of imaging and biodistribution indicated the highly specific accumulation of tumor after intravenous (i.v.) injection of (188)Re-liposome. The therapeutic efficacy of radiotherapeutics of (188)Re-liposome have been confirmed in a LS-174T solid tumor animal model, which points to the potential benefit and promise of passive nanoliposome delivered radiotherapeutics for cancer treatment.
Collapse
Affiliation(s)
- Chin-Wei Hsu
- Institute of Nuclear Energy Research , Taoyuan, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
13
|
Lo ST, Kumar A, Hsieh JT, Sun X. Dendrimer nanoscaffolds for potential theranostics of prostate cancer with a focus on radiochemistry. Mol Pharm 2013; 10:793-812. [PMID: 23294202 DOI: 10.1021/mp3005325] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Dendrimers are a class of structurally defined macromolecules featured with a central core, a low-density interior formed by repetitive branching units, and a high-density exterior terminated with surface functional groups. In contrast to their polymeric counterparts, dendrimers are nanosized and symmetrically shaped, which can be reproducibly synthesized on a large scale with monodispersity. These unique features have made dendrimers of increasing interest for drug delivery and other biomedical applications as nanoscaffold systems. Intended to address the potential use of dendrimers for the development of theranostic agents, which combines therapeutics and diagnostics in a single entity for personalized medicine, this review focuses on the reported methodologies of using dendrimer nanoscaffolds for targeted imaging and therapy of prostate cancer. Of particular interest, relevant chemistry strategies are discussed due to their important roles in the design and synthesis of diagnostic and therapeutic dendrimer-based nanoconjugates and potential theranostic agents, targeted or nontargeted. Given the developing status of nanoscaffolded theranostics, major challenges and potential hurdles are discussed along with the examples representing current advances.
Collapse
Affiliation(s)
- Su-Tang Lo
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | |
Collapse
|
14
|
Huang FYJ, Lee TW, Kao CHK, Chang CH, Zhang X, Lee WY, Chen WJ, Wang SC, Lo JM. Imaging, autoradiography, and biodistribution of (188)Re-labeled PEGylated nanoliposome in orthotopic glioma bearing rat model. Cancer Biother Radiopharm 2012; 26:717-25. [PMID: 22145660 DOI: 10.1089/cbr.2011.1052] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The (188)Re-labeled pegylated nanoliposome (abbreviated as (188)Re-Liposome) was prepared and evaluated for its potential as a theragnostic agent for glioma. (188)Re-BMEDA complex was loaded into the pegylated liposome core with pH 5.5 ammonium sulfate gradient to produce (188)Re-Liposome. Orthotopic Fischer344/F98 glioma tumor-bearing rats were prepared and intravenously injected with (188)Re-Liposome. Biodistribution, pharmacokinetic study, autoradiography (ARG), histopathology, and nano-SPECT/CT imaging were conducted for the animal model. The result showed that (188)Re-Liposome accumulated in the brain tumor of the animal model from 0.28%±0.09% injected dose (ID)/g (n=3) at 1 hour to a maximum of 1.95%±0.35% ID/g (n=3) at 24 hours postinjection. The tumor-to-normal brain uptake ratio (T/N ratio) increased from 3.5 at 1 hour to 32.5 at 24 hours. Both ARG and histopathological images clearly showed corresponding tumor regions with high T/N ratios. Nano-SPECT/CT detected a very clear tumor image from 4 hours till 48 hours. This study reveals the potential of (188)Re-Liposome as a theragnostic agent for brain glioma.
Collapse
Affiliation(s)
- Feng-Yun J Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Chi-Mou L, Chia-Che T, Chia-Yu Y, Wan-Chi L, Chung-Li H, Tsui-Jung C, Chih-Hsien C, Te-Wei L. Extended acute toxicity study of (188) Re-liposome in rats. J Appl Toxicol 2012; 33:886-93. [PMID: 22535684 DOI: 10.1002/jat.2751] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 02/23/2012] [Accepted: 02/23/2012] [Indexed: 11/10/2022]
Abstract
Liposomes can selectively target cancer sites and carry payloads, thereby improving diagnostic and therapeutic effectiveness as well as reducing toxicity. To evaluate therapeutic strategies, it is essential to use animal models reflecting important safety aspects before clinical application. As our previous study found that a high dosage (185 of MBq) of (188) Re-N,N-bis (2-mercaptoethyl)-N',N'-diethylethylenediamine-labeled pegylated liposomes ((188) Re-liposome) induced a decrease in white blood cell (WBC) count in Sprague-Dawley rats 7 days postinjection, the objective of the present study was to investigate extended acute radiotoxicity of (188) Re-liposome. Rats were administered via intravenous (i.v.) injection with (188) Re-liposome (185, 55.5 and 18.5 MBq), normal saline as a blank control or non-radioactive liposome as a vehicle control. Mortality, clinical signs, food consumption, body weights, urinary, biochemical and hematological analyzes were examined. In addition, gross necropsy and histopathological examinations were also performed at the end of the follow-up period. None of the rats died and no clinical sign was observed during the 28-day study period. Only male rats receiving (188) Re-liposome at a high dosage (185 MBq) displayed a slight weight loss compared with the control rats. In both male and female rats, the WBC counts of both high-dose and medium-dose (55.5 MBq) groups reduced significantly 7 days postinjection, but recovered to the normal range on Study Day 29. There was no significant difference in urinary analyzes, biochemical parameters and histopathological assessments between the (188) Re-liposome-treated and control groups. The information generated from the present study on extended acute toxicity of (188) Re-liposome will serve as a safety reference for radiopharmaceuticals in early-phase clinical trials.
Collapse
Affiliation(s)
- Liu Chi-Mou
- Institute of Nuclear Energy Research, Taoyuan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Chen LC, Wu YH, Liu IH, Ho CL, Lee WC, Chang CH, Lan KL, Ting G, Lee TW, Shien JH. Pharmacokinetics, dosimetry and comparative efficacy of 188Re-liposome and 5-FU in a CT26-luc lung-metastatic mice model. Nucl Med Biol 2012; 39:35-43. [DOI: 10.1016/j.nucmedbio.2011.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 06/13/2011] [Accepted: 06/21/2011] [Indexed: 01/19/2023]
|
17
|
Lin YY, Chang CH, Li JJ, Stabin MG, Chang YJ, Chen LC, Lin MH, Tseng YL, Lin WJ, Lee TW, Ting G, Chang CA, Chen FD, Wang HE. Pharmacokinetics and dosimetry of (111)In/(188)Re-labeled PEGylated liposomal drugs in two colon carcinoma-bearing mouse models. Cancer Biother Radiopharm 2011; 26:373-80. [PMID: 21711113 DOI: 10.1089/cbr.2010.0906] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PEGylated liposomes are important drug carriers for nanomedicine cancer therapy. PEGylated liposomes can encapsulate radio- and chemo-drugs and passively target tumor sites via enhanced permeability and retention effect. This study estimated the pharmacokinetics and dosimetry after administration of radio-chemotherapeutics ((111)In-labeled vinorelbine [VNB]-encapsulated liposomes, InVNBL, and (188)Re-labeled doxorubicin [DXR]-encapsulated liposomes, ReDXRL) for radionuclide therapy in two colon carcinoma-bearing mouse models. A C26 colon carcinoma tumor/ascites mouse model and a subcutaneous solid tumor-bearing mouse model were employed. Biodistribution studies of InVNBL and ReDXRL after intraperitoneal administration in tumor/ascites-bearing mice (protocol A) and intravenous administration in subcutaneous solid tumor-bearing mice (protocol B) were performed. The radiation dose to normal tissues and tumors were calculated based on the results of distribution studies in mice, using the OLINDA/EXM program. The cumulated activities in most organs after administration of InVNBL in either the tumor/ascites-bearing mice (protocol A) or the subcutaneous solid tumor-bearing mice (protocol B) were higher than those of ReDXRL. Higher tumor-to-normal-tissues absorption dose ratios (T/NTs) were observed after administration of InVNBL than those of ReDXRL for protocol A. The T/NTs for the liver, spleen, and red marrow after injection of InVNBL for protocol B were similar to those of ReDXRL. The critical organ was found to be red marrow, and thus the red marrow absorption dose defined the recommended maximum administration activity of these liposomal drugs. Characterization of pharmacokinetics and dosimetry is needed to select the appropriate radiotherapeutics for specific tumor treatment applications. The results suggest that InVNBL is a promising therapeutic agent, which is as good as ReDXRL, in two mouse tumor models.
Collapse
Affiliation(s)
- Yi-Yu Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Local delivery of rhenium-188 colloid into hepatic tumor sites in rats using thermo-sensitive chitosan hydrogel: effects of gelling time of chitosan as delivery system. J Radioanal Nucl Chem 2011. [DOI: 10.1007/s10967-011-1111-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
19
|
Chang CH, Chiu SP, Chiang TC, Lee TW. Acute intravenous injection toxicity of BMEDA in mice. Drug Chem Toxicol 2011; 34:20-4. [PMID: 21121875 DOI: 10.3109/01480545.2010.482588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
(188)Re/(186)Re-N,N-bis(2-mercaptoethyl)-N',N'-diethylethylenediamine-labeled pegylated liposome ((188)Re-BMEDA-liposome) has been proven as a promising candidate for cancer therapy in tumor-rodent models. (188)Re-BMEDA complexes should be prepared for the radiolabeling of liposomes. This article describes the acute toxicity of BMEDA in Imprinting Control Region (ICR) mice. Treated mice were administered with BMEDA at dose levels of 3, 6, 9, and 12 mg/kg, with a dose volume of 10 mL/kg. The control mice were administered 10 mL/kg of vehicle control. The mice were observed for 14 days. Observations included mortality, clinical signs, total body-weight gains, food consumption, and gross necropsy findings. BMEDA exerted no adverse toxic effects in ICR mice at dose levels 3 mg/kg, which are up to 360,000 times higher than the intended human dose. The lethal-dose (LD(50)) value of BMEDA was 8.13 and 8.68 mg/kg in male and female mice, respectively.
Collapse
Affiliation(s)
- Chih-Hsien Chang
- Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan, Taiwan
| | | | | | | |
Collapse
|
20
|
Liu CM, Chang CH, Chang YJ, Hsu CW, Chen LC, Chen HL, Ho CL, Yu CY, Chang TJ, Chiang TC, Lee TW. Preliminary evaluation of acute toxicity of (188) Re-BMEDA-liposome in rats. J Appl Toxicol 2011; 30:680-7. [PMID: 20981861 DOI: 10.1002/jat.1541] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Liposomes can selectively target cancer sites and carry payloads, thereby improving diagnostic and therapeutic effectiveness and reducing toxicity. To evaluate therapeutic strategies, it is essential to use animal models reflecting important safety aspects before clinical application. The objective of this study was to investigate acute radiotoxicity of ¹⁸⁸Re-N,N-bis (2-mercaptoethyl)-N',N'-diethylethylenediamine (BMEDA)-labeled pegylated liposomes (¹⁸⁸Re-BMEDA-liposome) in Sprague-Dawley rats. Rats were administered with ¹⁸⁸Re-BMEDA-liposome, normal saline as blank or non-radioactive liposome as vehicle control via intravenous injection and observed for 14 days. Examinations were conducted with respect to mortality, clinical signs, food consumption, body weight and hematological and biochemical analyses. In addition, gross necropsy, histopathological examinations and cytogenetic analyses were also performed. None of the rats died and no clinical sign was observed during the 14-day study period. Rats administered with ¹⁸⁸Re-BMEDA-liposome at dosage of 185 MBq displayed a significant weight loss compared with the control from study day (SD) 1 to SD 4, and the white blood cell count reduced to 5-10% of initial value (female: 18.55 ± 6.58 to 0.73 ± 0.26 x 10³ µl⁻¹; male: 14.52 ± 5.12 to 1.43 ± 0.54 x 10³ µl⁻¹) 7 days-post injection, but were found to have recovered on SD 15. There were no significant differences in biochemical parameters and histopathological assessments between the ¹⁸⁸Re-BMEDA-liposome-treated and control groups. The frequencies of dicentric chromosomes were associated with dosage of ¹⁸⁸Re-BMEDA-liposome. The information generated from this study on acute toxicity will serve as a safety reference for further subacute toxicity study in rats and human clinical trials.
Collapse
Affiliation(s)
- Chi-Mou Liu
- Institute of Nuclear Energy Research, Tauyuan, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Nanotargeted radionuclides for cancer nuclear imaging and internal radiotherapy. J Biomed Biotechnol 2010; 2010. [PMID: 20811605 PMCID: PMC2929518 DOI: 10.1155/2010/953537] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 06/15/2010] [Indexed: 12/18/2022] Open
Abstract
Current progress in nanomedicine has exploited the possibility of designing tumor-targeted nanocarriers being able to deliver radionuclide payloads in a site or molecular selective manner to improve the efficacy and safety of cancer imaging and therapy. Radionuclides of auger electron-, α-, β-, and γ-radiation emitters have been surface-bioconjugated or after-loaded in nanoparticles to improve the efficacy and reduce the toxicity of cancer imaging and therapy in preclinical and clinical studies. This article provides a brief overview of current status of applications, advantages, problems, up-to-date research and development, and future prospects of nanotargeted radionuclides in cancer nuclear imaging and radiotherapy. Passive and active nanotargeting delivery of radionuclides with illustrating examples for tumor imaging and therapy are reviewed and summarized. Research on combing different modes of selective delivery of radionuclides through nanocarriers targeted delivery for tumor imaging and therapy offers the new possibility of large increases in cancer diagnostic efficacy and therapeutic index. However, further efforts and challenges in preclinical and clinical efficacy and toxicity studies are required to translate those advanced technologies to the clinical applications for cancer patients.
Collapse
|
22
|
Joshi BP, Wang TD. Exogenous Molecular Probes for Targeted Imaging in Cancer: Focus on Multi-modal Imaging. Cancers (Basel) 2010; 2:1251-87. [PMID: 22180839 PMCID: PMC3237638 DOI: 10.3390/cancers2021251] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 05/31/2010] [Accepted: 06/02/2010] [Indexed: 01/09/2023] Open
Abstract
Cancer is one of the major causes of mortality and morbidity in our health care system. Molecular imaging is an emerging methodology for the early detection of cancer, and the development of exogenous molecular probes that can be labeled for multi-modality imaging is critical to this process. Today, molecular imaging is at crossroad, and new targeted imaging agents are expected to broadly expand our ability to detect pre-malignant lesions. This integrated imaging strategy will permit clinicians to not only localize lesions within the body, but also to visualize the expression and activity of specific molecules. This information is expected to have a major impact on diagnosis, therapy, drug development and understanding of basic cancer biology. At this time, a number of molecular probes have been developed by conjugating various labels to affinity ligands for targeting in different imaging modalities. This review will describe the current status of exogenous molecular probes for optical, nuclear and MRI imaging platforms. Furthermore, we will also shed light on how these techniques can be used synergistically in multi-modal platforms and how these techniques are being employed in current research.
Collapse
Affiliation(s)
- Bishnu P. Joshi
- Division of Gastroenterology, Department of Medicine, University of Michigan, School of Medicine, 109 Zina Pitcher Place, BSRB 1722, Ann Arbor, MI 48109, USA
| | - Thomas D. Wang
- Division of Gastroenterology, Department of Medicine, University of Michigan, School of Medicine, 109 Zina Pitcher Place, BSRB 1722, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
23
|
Lin YY, Li JJ, Chang CH, Lu YC, Hwang JJ, Tseng YL, Lin WJ, Ting G, Wang HE. Evaluation of pharmacokinetics of 111In-labeled VNB-PEGylated liposomes after intraperitoneal and intravenous administration in a tumor/ascites mouse model. Cancer Biother Radiopharm 2009; 24:453-60. [PMID: 19694580 DOI: 10.1089/cbr.2008.0572] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Nanoliposomes are important drug carriers that can passively target tumor sites by the enhanced permeability and retention (EPR) effect in neoplasm lesions. This study evaluated the biodistribution and pharmacokinetics of 111In-labeled vinorelbine (VNB)-encapsulated PEGylated liposomes (IVNBPL) after intraperitoneal (i.p.) and intravenous (i.v.) administration in a C26/tk-luc colon carcinoma ascites mouse model. IVNBPL was prepared by labeling VNB-encapsulated PEGylated liposomes with 111In-oxine. BALB/c mice were i.p. inoculated with 2 x 10(5) C26/tk-luc cells in 500 muL of phosphate-buffered saline. Peritoneal tumor lesions were confirmed by 124I-FIAU/micro-PET (positron emission tomography) and bioluminescence imaging. Ascites production was examined by ultrasound imaging on day 10 after tumor cell inoculation. The pharmacokinetics and biodistribution studies of IVNBPL in a tumor/ascites mouse model were conducted. The labeling efficiency was more than 90%. The in vitro stability in human plasma at 37 degrees C for 72 hours was 83% +/- 3.5%. For i.p. administration, the areas under curves (AUCs) of ascites and tumor were 6.78- and 1.70-fold higher, whereas the AUCs of normal tissues were lower than those via the i.v. route. This study demonstrates that i.p. administration is a better approach than i.v. injection for IVNBPL, when applied to the treatment of i.p. malignant disease in a tumor/ascites mouse model.
Collapse
Affiliation(s)
- Yi-Yu Lin
- Biomedical Imaging and Radiological Sciences, National Yang-Ming University , Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chen LC, Chang CH, Yu CY, Chang YJ, Wu YH, Lee WC, Yeh CH, Lee TW, Ting G. Pharmacokinetics, micro-SPECT/CT imaging and therapeutic efficacy of (188)Re-DXR-liposome in C26 colon carcinoma ascites mice model. Nucl Med Biol 2009; 35:883-93. [PMID: 19026950 DOI: 10.1016/j.nucmedbio.2008.09.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 08/25/2008] [Accepted: 09/13/2008] [Indexed: 02/04/2023]
Abstract
The pharmacokinetics and internal radionuclide therapy of intraperitoneally administrated (188)Re-N,N-bis(2-mercaptoethyl)-N',N'-diethylethylenediamine (BMEDA)-labeled pegylated liposomal doxorubicin ((188)Re-DXR-liposome) were investigated in the C26 murine colon carcinoma ascites mouse model. After intraperitoneal administration of the nanotargeted bimodality (188)Re-DXR-liposome, the ascites and tumor accumulation of the radioactivity were observed, the levels of radioactivity within the ascites were maintained at relatively higher levels before 48 h and the levels of radioactivity in the tumor were maintained at steady levels after 4 h. The AUC((o-->infinity)) of (188)Re-DXR-liposome in blood, ascites and tumor was 9.3-, 4.2- and 4.7-fold larger than that of (188)Re-BMEDA, respectively. The maximum tolerated dose of intraperitoneally administrated (188)Re-DXR-liposome was determined in normal BALB/c mice. The survival, tumor and ascites inhibition of mice after (188)Re-DXR-liposome (22.2 MBq of (188)Re, 5 mg/kg of DXR) treatment were evaluated. Consequently, radiochemotherapeutics of (188)Re-DXR-liposome attained better survival time, tumor and ascites inhibition (decreased by 49% and 91% at 4 days after treatment; P<.05) in mice than radiotherapeutics of (188)Re-liposome or chemotherapeutics of Lipo-Dox did. Therefore, intraperitoneal administration of novel (188)Re-DXR-liposome could provide a benefit and promising strategy for delivery of passive nanotargeted bimodality radiochemotherapeutics in oncology applications.
Collapse
|