1
|
Kim E, Lee C, Seo Y, Hong D, Song B, Kim J, Jeong W, Kwon SY, Kim DY, Pyo A. Synthesis and evaluation of a 68Ga-labeled iodinated benzamide derivative as a PET imaging agent for malignant melanoma. Appl Radiat Isot 2025; 220:111791. [PMID: 40127585 DOI: 10.1016/j.apradiso.2025.111791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Malignant melanoma is a highly aggressive skin cancer with increasing prevalence worldwide. The 5-year survival rate for localized malignant melanoma is 90%, but this drops to 6% if metastasis has occurred at diagnosis. Current positron emission tomography (PET) imaging probes, such as 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG), show low sensitivity for metastatic melanoma, underscoring the need for new probes. This study explores the potential of 68Ga-labeled 2,2',2″,2‴-(2-(4-(3-(2-((2-(5-iodopicolinamido)ethyl)(methyl)amino)ethyl)thioureido)benzyl)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetrayl)tetraacetic acid (68Ga-MI-0202F1) as a novel PET imaging probe designed for enhanced specificity and sensitivity towards melanin. The precursor MI-0202F1 was synthesized from 5-iodopicolinic acid, 2,2'-diamino-N-methyldiethylamine, and 2,2',2″,2‴-(2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetrayl)tetraacetic acid ((p-SCN-Bn)-DOTA). The precursor was labeled with 68Ga in a reaction vial at 95°C and pH 5.0 for 10 min. The radiochemical purity of the 68Ga-labeled compound was determined using radio-thin-layer chromatography. Cellular uptake of the iodinated benzamide derivative was evaluated in the B16F10 cell line (mouse melanoma) following L-tyrosine treatment. MicroPET studies were conducted at 30 and 60 min post-intravenous injection of the labeled complex into B16F10 tumor-bearing mice. The non-decay-corrected radiochemical yield was 95.96 ± 3.73% (n = 3), and the radiochemical purity exceeded 98%. In vitro studies demonstrated significantly higher uptake of 68Ga-MI-0202F1 in L-tyrosine-treated B16F10 cells compared with untreated cells. In vivo PET imaging showed rapid accumulation and sustained retention of 68Ga-MI-0202F1 in melanoma tumors, with high tumor-to-background ratios. Overall, 68Ga-MI-0202F1 demonstrates potential as an innovative molecular imaging probe for malignant melanoma. Further studies are needed to investigate the potential of MI-0202F1 as a theranostic agent incorporating the radioisotope pairs 68Ga and 177Lu.
Collapse
Affiliation(s)
- Eunsu Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Chaewon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Yejin Seo
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Dagyeong Hong
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Boreum Song
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Jiyu Kim
- CNCure Biotech, Hwasun, Republic of Korea
| | | | - Seong-Young Kwon
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Dong-Yeon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea; CNCure Biotech, Hwasun, Republic of Korea.
| | - Ayoung Pyo
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea.
| |
Collapse
|
2
|
Tao J, Cao R, Lei Z, Wang X, Luo R, Zhou Y, Li Y, Xu X, Liu X, Qu C, Song S, Tian M, Cheng Z. Development of 99mTc-labeled melanin-targeted probes for SPECT imaging of melanoma. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07268-1. [PMID: 40257608 DOI: 10.1007/s00259-025-07268-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/03/2025] [Indexed: 04/22/2025]
Abstract
PURPOSE Melanoma is a highly aggressive skin cancer with poor prognosis, highlighting the need for early detection. This study aims to develop a 99mTc-labeled single photon emission computed tomography (SPECT) probe targeting melanin to improve melanoma detection with enhanced specificity and cost-effectiveness. METHODS Six potential 99mTc-labeled probes were synthesized by conjugating small molecules with nitrogen-rich fatty chains or cyclic structures to 99mTc. These 99mTc-complexes were then assessed in vitro for their lipid-water partition coefficient, radiochemical stability, and cellular uptake. In vivo studies included biodistribution, SPECT/CT imaging, pharmacokinetics, stability and biocompatibility studies were conducted in various mouse models. RESULTS These six probes exhibited high radiolabeling yields (> 95%) and radiochemical purities (> 95%). In vitro cell uptake and blocking experiments demonstrated their binding capability and specificity for melanin. Among these probes, 99mTc-SMIC-4006 displayed the highest tumor uptake (5.18 ± 1.55%ID/g at 1 h) and significantly higher uptake in B16F10 xenografts compared to non-melanotic A375 xenografts (p < 0.01). The initial tumor-to-blood ratio for 99mTc-SMIC-4006 was 4.30 ± 0.63 at 1 h, increasing to 10.27 ± 5.13 at 6 h. Importantly, SPECT/CT imaging of 99mTc-SMIC-4006 clearly delineated B16F10 tumor regions with high contrast, while A375 xenografts showed minimal uptake. CONCLUSION 99mTc-SMIC-4006 is a promising SPECT probe for melanoma imaging, offering high tumor specificity, excellent uptake, and ideal tumor-to-background contrast, with strong potential for clinical application.
Collapse
Affiliation(s)
- Ji Tao
- Human Phenome Institute, Fudan University, Shanghai, China
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Rui Cao
- Department of Nuclear Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhe Lei
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao Wang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Renli Luo
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyuan Zhou
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yanlin Li
- Human Phenome Institute, Fudan University, Shanghai, China
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaosheng Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chunrong Qu
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Mei Tian
- Human Phenome Institute, Fudan University, Shanghai, China.
- Department of Nuclear Medicine/PET Center, Huashan Hospital, Fudan University, Shanghai, China.
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China.
| |
Collapse
|
3
|
Bunda S, Kálmán-Szabó I, Lihi N, Képes Z, Szikra D, Peline Szabo J, Timári I, Szücs D, May NV, Papp G, Trencsényi G, Kálmán FK. Diagnosis of Melanoma with 61Cu-Labeled PET Tracer. J Med Chem 2024; 67:9342-9354. [PMID: 38753457 DOI: 10.1021/acs.jmedchem.4c00479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Until the recent years, substances containing radioactive 61Cu were strongly considered as potential positron-emitting radiopharmaceuticals for use in positron emission tomography (PET) applications; however, due to their suitably long half-life, and generator-independent and cost-effective production, they seem to be economically viable for human imaging. Since malignant melanoma (MM) is a major public health problem, its early diagnosis is a crucial contributor to long-term survival, which can be achieved using radiolabeled α-melanocyte-stimulating hormone analog NAPamide derivatives. Here, we report on the physicochemical features of a new CB-15aneN5-based Cu(II) complex ([Cu(KFTGdiac)]-) and the ex vivo and in vivo characterization of its NAPamide conjugate. The rigid chelate possesses prompt complex formation and suitable inertness (t1/2 = 18.4 min in 5.0 M HCl at 50 °C), as well as excellent features in the diagnosis of B16-F10 melanoma tumors (T/M(SUVs) (in vivo): 12.7, %ID/g: 6.6 ± 0.3, T/M (ex vivo): 22).
Collapse
Affiliation(s)
- Szilvia Bunda
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| | - Ibolya Kálmán-Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Norbert Lihi
- HUN-REN-UD Mechanisms of Complex Homogeneous and Heterogeneous Chemical Reactions Research Group, Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Dezső Szikra
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Judit Peline Szabo
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - István Timári
- Department of Organic Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- HUN-REN-UD Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Dániel Szücs
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Nóra V May
- Centre for Structural Science, Research Centre for Natural Sciences, Hungarian Research Network (HUN-REN), Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Gábor Papp
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Ferenc K Kálmán
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| |
Collapse
|
4
|
Zhang X, Lin Z, Feng Y, Kang F, Wang J, Lan X. Melanin-Targeting Radiotracers and Their Preclinical, Translational, and Clinical Status: From Past to Future. J Nucl Med 2024; 65:19S-28S. [PMID: 38719238 DOI: 10.2967/jnumed.123.266945] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/31/2024] [Indexed: 07/16/2024] Open
Abstract
Melanin is one of the representative biomarkers of malignant melanoma and a potential target for diagnosis and therapy. With advancements in chemistry and radiolabeling technologies, promising strides have been made to synthesize radiolabeled melanin-binding molecules for various applications. We present an overview of melanin-targeted radiolabeled molecules and compare their features reported in preclinical studies. Clinical practice and trials are also discussed to elaborate on the safety and validity of the probes, and expanded applications beyond melanoma are reviewed. Melanin-targeted imaging holds potential value in the diagnosis, staging, and prognostic assessment of melanoma and other applications. Melanin-targeted radionuclide therapy possesses immense potential but requires more clinical validation. Furthermore, an intriguing avenue for future research involves expanding the application scope of melanin-targeted probes and exploring their value.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, Ministry of Education, Wuhan, China; and
| | - Zhaoguo Lin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, Ministry of Education, Wuhan, China; and
| | - Yuan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, Ministry of Education, Wuhan, China; and
| | - Fei Kang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, Ministry of Education, Wuhan, China; and
| |
Collapse
|
5
|
Pyo A, Yun M, Song B, Kwon SY, Min JJ, Kim DY. Synthesis and evaluation of 18F-labeled procainamide as a PET imaging agent for malignant melanoma. Bioorg Med Chem Lett 2023; 96:129528. [PMID: 37852422 DOI: 10.1016/j.bmcl.2023.129528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/14/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Malignant melanoma has an aggressive nature and a high metastatic propensity resulting in the highest mortality rate of any skin cancer. In this study, we synthesized 18F-labeled procainamide (PCA) for detection of melanoma using positron emission tomography (PET), and evaluated its biological characteristics. The non-decay-corrected radiochemical yield of 18F-PCA was 10-15% and its in vitro stability was over 98% for 2 h. At 1 h, cellular uptake of 18F-PCA was 3.8-fold higher in a group with the presence of l-tyrosine than in a non-l-tyrosine-treated group. Furthermore, 18F-PCA permitted visualization of B16F10 (mouse melanoma) xenografts on microPET after intravenous injection, and was retained in the tumor for 60 min, with a high tumor-to-liver uptake ratio. 18F-PCA showed specific melanoma uptake in primary lesions with a high melanin targeting ability in small animal models. 18F-PCA may have potential as a PET imaging agent for direct melanoma detection.
Collapse
Affiliation(s)
- Ayoung Pyo
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Misun Yun
- Hygenic Safety-Material Research Group, Technology Innovation Research Division, World Institute of Kimchi, Gwangju, Republic of Korea
| | - Boreum Song
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Seong-Young Kwon
- Innovation Center for Molecular Probe Development, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Jung-Joon Min
- Innovation Center for Molecular Probe Development, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea; CNCure Biotech, Hwasun, Republic of Korea
| | - Dong-Yeon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, Republic of Korea; CNCure Biotech, Hwasun, Republic of Korea.
| |
Collapse
|
6
|
Shi H, Cheng Z. MC1R and melanin-based molecular probes for theranostic of melanoma and beyond. Acta Pharmacol Sin 2022; 43:3034-3044. [PMID: 36008707 PMCID: PMC9712491 DOI: 10.1038/s41401-022-00970-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/27/2022] [Indexed: 12/12/2022]
Abstract
Malignant melanoma is accounting for most of skin cancer-associated mortality. The incidence of melanoma increased every year worldwide especially in western countries. Treatment efficiency is highly related to the stage of melanoma. Therefore, accurate staging and restaging play a pivotal role in the management of melanoma patients. Though 18F-fluorodeoxyglucose (18F-FDG) positron-emission tomography (PET) has been widely used in imaging of tumor metastases, novel radioactive probes for specific targeted imaging of both primary and metastasized melanoma are still desired. Melanocortin receptor 1 (MC1R) and melanin are two promising biomarkers specifically for melanoma, and numerous research groups including us have been actively developing a plethora of radioactive probes based on targeting of MC1R or melanin for over two decades. In this review, some of the MC1R-targeted tracers and melanin-associated molecular imaging probes developed in our research and others have been briefly summarized, and it provides a quick glance of melanoma-targeted probe design and may contribute to further developing novel molecular probes for cancer theranostics.
Collapse
Affiliation(s)
- Hui Shi
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
7
|
Li T, Yang Y, Jing W, Yan Z, Che J, Xu H, Hu X, Zhang R. Melanin-gelatin nanoparticles with both EPR effect and renal clearance for PA/MRI dual-modal imaging of tumors. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 134:112718. [DOI: 10.1016/j.msec.2022.112718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/31/2022] [Accepted: 02/12/2022] [Indexed: 01/09/2023]
|
8
|
Han HS, Koo SY, Choi KY. Emerging nanoformulation strategies for phytocompounds and applications from drug delivery to phototherapy to imaging. Bioact Mater 2021; 14:182-205. [PMID: 35310344 PMCID: PMC8892098 DOI: 10.1016/j.bioactmat.2021.11.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
Over thousands of years, natural bioactive compounds derived from plants (bioactive phytocompounds, BPCs) have been used worldwide to address human health issues. Today, they are a significant resource for drug discovery in the development of modern medicines. Although many BPCs have promising biological activities, most of them cannot be effectively utilized in drugs for therapeutic applications because of their inherent limitations of low solubility, structural instability, short half-life, poor bioavailability, and non-specific distribution to organs. Researchers have utilized emerging nanoformulation (NF) technologies to overcome these limitations as they have demonstrated great potential to improve the solubility, stability, and pharmacokinetic and pharmacodynamic characteristics of BPCs. This review exemplifies NF strategies for resolving the issues associated with BPCs and summarizes recent advances in their preclinical and clinical applications for imaging and therapy. This review also highlights how innovative NF technologies play a leading role in next-generation BPC-based drug development for extended therapeutic applications. Finally, this review discusses the opportunities to take BPCs with meaningful clinical impact from bench to bedside and extend the patent life of BPC-based medicines with new formulations or application to new adjacent diseases beyond the primary drug indications. Natural bioactive phytocompounds derived from plants have been used worldwide to address human health issues. However, most of them cannot be effectively utilized in drugs for therapeutic applications because of their inherent limitations. Nanoformulation approach has recently been underlined as an emerging pharmaceutical strategy to overcome the intrinsic drawbacks of bioactive phytocompounds. Various types of nanoformulation and their up-to-date applications for targeted delivery, phototherapy, and imaging are reviewed. Finally, their clinical implications for the repurposing of bioactive phytocompounds are deliberated.
Collapse
Affiliation(s)
- Hwa Seung Han
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Song Yi Koo
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Ki Young Choi
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- Corresponding author. Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea.
| |
Collapse
|
9
|
Katz SR, Yakovlev MA, Vanselow DJ, Ding Y, Lin AY, Parkinson DY, Wang Y, Canfield VA, Ang KC, Cheng KC. Whole-organism 3D quantitative characterization of zebrafish melanin by silver deposition micro-CT. eLife 2021; 10:e68920. [PMID: 34528510 PMCID: PMC8445617 DOI: 10.7554/elife.68920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/19/2021] [Indexed: 01/10/2023] Open
Abstract
We previously described X-ray histotomography, a high-resolution, non-destructive form of X-ray microtomography (micro-CT) imaging customized for three-dimensional (3D), digital histology, allowing quantitative, volumetric tissue and organismal phenotyping (Ding et al., 2019). Here, we have combined micro-CT with a novel application of ionic silver staining to characterize melanin distribution in whole zebrafish larvae. The resulting images enabled whole-body, computational analyses of regional melanin content and morphology. Normalized micro-CT reconstructions of silver-stained fish consistently reproduced pigment patterns seen by light microscopy, and further allowed direct quantitative comparisons of melanin content across wild-type and mutant samples, including subtle phenotypes not previously noticed. Silver staining of melanin for micro-CT provides proof-of-principle for whole-body, 3D computational phenomic analysis of a specific cell type at cellular resolution, with potential applications in other model organisms and melanocytic neoplasms. Advances such as this in whole-organism, high-resolution phenotyping provide superior context for studying the phenotypic effects of genetic, disease, and environmental variables.
Collapse
Affiliation(s)
- Spencer R Katz
- Division of Experimental Pathology, Department of Pathology, Pennsylvania State University College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
- Medical Scientist Training Program, Penn State College of MedicineHersheyUnited States
| | - Maksim A Yakovlev
- Division of Experimental Pathology, Department of Pathology, Pennsylvania State University College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
| | - Daniel J Vanselow
- Division of Experimental Pathology, Department of Pathology, Pennsylvania State University College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
| | - Yifu Ding
- Division of Experimental Pathology, Department of Pathology, Pennsylvania State University College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
- Medical Scientist Training Program, Penn State College of MedicineHersheyUnited States
| | - Alex Y Lin
- Division of Experimental Pathology, Department of Pathology, Pennsylvania State University College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
| | | | - Yuxin Wang
- Mobile Imaging Innovations, IncPalatineUnited States
| | - Victor A Canfield
- Division of Experimental Pathology, Department of Pathology, Pennsylvania State University College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
| | - Khai C Ang
- Division of Experimental Pathology, Department of Pathology, Pennsylvania State University College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
- Zebrafish Functional Genomics Core, Penn State College of MedicineHersheyUnited States
| | - Keith C Cheng
- Division of Experimental Pathology, Department of Pathology, Pennsylvania State University College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
- Zebrafish Functional Genomics Core, Penn State College of MedicineHersheyUnited States
| |
Collapse
|
10
|
Rouanet J, Quintana M, Auzeloux P, Cachin F, Degoul F. Benzamide derivative radiotracers targeting melanin for melanoma imaging and therapy: Preclinical/clinical development and combination with other treatments. Pharmacol Ther 2021; 224:107829. [PMID: 33662452 DOI: 10.1016/j.pharmthera.2021.107829] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 12/16/2022]
Abstract
Cutaneous melanoma arises from proliferating melanocytes, cells specialized in the production of melanin. This property means melanin can be considered as a target for monitoring melanoma patients using nuclear imaging or targeted radionuclide therapy (TRT). Since the 1970s, many researchers have shown that specific molecules can interfere with melanin. This paper reviews some such molecules: benzamide structures improved to increase their pharmacokinetics for imaging or TRT. We first describe the characteristics and biosynthesis of melanin, and the main features of melanin tracers. The second part summarizes the preclinical and corresponding clinical studies on imaging. The last section presents TRT results from ongoing protocols and discusses combinations with other therapies as an opportunity for melanoma non-responders or patients resistant to treatments.
Collapse
Affiliation(s)
- Jacques Rouanet
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, 58 Rue Montalembert, 63005 Clermont-Ferrand, Cedex, France; Department of Dermatology and Oncodermatology, CHU Estaing, 1 place Lucie et Raymond Aubrac, 63000 Clermont-Ferrand, France; Centre Jean Perrin, Clermont-Ferrand F-63011, France.
| | - Mercedes Quintana
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, 58 Rue Montalembert, 63005 Clermont-Ferrand, Cedex, France.
| | - Philippe Auzeloux
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, 58 Rue Montalembert, 63005 Clermont-Ferrand, Cedex, France.
| | - Florent Cachin
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, 58 Rue Montalembert, 63005 Clermont-Ferrand, Cedex, France; Centre Jean Perrin, Clermont-Ferrand F-63011, France.
| | - Françoise Degoul
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, 58 Rue Montalembert, 63005 Clermont-Ferrand, Cedex, France.
| |
Collapse
|
11
|
Chen CC, Chen YY, Lo YH, Lin MH, Chang CH, Chen CL, Wang HE, Wu CY. Evaluation of Radioiodinated Fluoronicotinamide/Fluoropicolinamide-Benzamide Derivatives as Theranostic Agents for Melanoma. Int J Mol Sci 2020; 21:ijms21186597. [PMID: 32916962 PMCID: PMC7554940 DOI: 10.3390/ijms21186597] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/01/2022] Open
Abstract
Malignant melanoma is the most harmful type of skin cancer and its incidence has increased in this past decade. Early diagnosis and treatment are urgently desired. In this study, we conjugated picolinamide/nicotinamide with the pharmacophore of 131I-MIP-1145 to develop 131I-iodofluoropicolinamide benzamide (131I-IFPABZA) and 131I-iodofluoronicotiamide benzamide (131I-IFNABZA) with acceptable radiochemical yield (40 ± 5%) and high radiochemical purity (>98%). We also presented their biological characteristics in melanoma-bearing mouse models. 131I-IFPABZA (Log P = 2.01) was more lipophilic than 131I-IFNABZA (Log P = 1.49). B16F10-bearing mice injected with 131I-IFNABZA exhibited higher tumor-to-muscle ratio (T/M) than those administered with 131I-IFPABZA in planar γ-imaging and biodistribution studies. However, the imaging of 131I-IFNABZA- and 131I-IFPABZA-injected mice only showed marginal tumor uptake in A375 amelanotic melanoma-bearing mice throughout the experiment period, indicating the high binding affinity of these two radiotracers to melanin. Comparing the radiation-absorbed dose of 131I-IFNABZA with the melanin-targeted agents reported in the literature, 131I-IFNABZA exerts lower doses to normal tissues on the basis of similar tumor dose. Based on the in vitro and in vivo studies, we clearly demonstrated the potential of using 131I-IFNABZA as a theranostic agent against melanoma.
Collapse
Affiliation(s)
- Chao-Cheng Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.C.); (Y.-Y.C.); (Y.-H.L.); (C.-H.C.); (C.-L.C.); (H.-E.W.)
| | - Yang-Yi Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.C.); (Y.-Y.C.); (Y.-H.L.); (C.-H.C.); (C.-L.C.); (H.-E.W.)
| | - Yi-Hsuan Lo
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.C.); (Y.-Y.C.); (Y.-H.L.); (C.-H.C.); (C.-L.C.); (H.-E.W.)
| | - Ming-Hsien Lin
- Department of Nuclear Medicine, Taipei City Hospital Zhongxiao Branch, Taipei 115, Taiwan;
- Department of Nuclear Medicine, Cheng Hsin General Hospital, Taipei 112, Taiwan
| | - Chih-Hsien Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.C.); (Y.-Y.C.); (Y.-H.L.); (C.-H.C.); (C.-L.C.); (H.-E.W.)
- Institute of Nuclear Energy Research, Taoyuan 325, Taiwan
| | - Chuan-Lin Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.C.); (Y.-Y.C.); (Y.-H.L.); (C.-H.C.); (C.-L.C.); (H.-E.W.)
| | - Hsin-Ell Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.C.); (Y.-Y.C.); (Y.-H.L.); (C.-H.C.); (C.-L.C.); (H.-E.W.)
| | - Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.C.); (Y.-Y.C.); (Y.-H.L.); (C.-H.C.); (C.-L.C.); (H.-E.W.)
- Correspondence:
| |
Collapse
|
12
|
Abstract
With the emergence of new therapeutic modalities, the diagnosis of melanoma at the earliest practicable stage has become more important for improving the survival of patients. We developed a positron emission tomography (PET) imaging probe, N-(2-(dimethylamino)ethyl)-5-[18F]fluoropicolinamide ([18F]DMPY2) and evaluated diagnostic performance in animal models. [18F]DMPY2 PET exhibited excellent performance in detecting primary and metastatic melanomas, demonstrating strong/prolonged tumoral uptake and rapid background clearance. This suggests that this radiotracer could be used as a novel PET imaging agent to obtain outstanding image quality in the diagnosis of melanoma. This is the pioneering report of pyridine-based benzamide derivative with reduced alkyl chains in the amine residue and ultrasensitive detection of melanoma lesions in living subjects compared to conventional PET imaging agents. Malignant melanoma has one of the highest mortality rates of any cancer because of its aggressive nature and high metastatic potential. Clinical staging of the disease at the time of diagnosis is very important for the prognosis and outcome of melanoma treatment. In this study, we designed and synthesized the 18F-labeled pyridine-based benzamide derivatives N-(2-(dimethylamino)ethyl)-5-[18F]fluoropicolinamide ([18F]DMPY2) and N-(2-(dimethylamino)ethyl)-6-[18F]fluoronicotinamide ([18F]DMPY3) to detect primary and metastatic melanoma at an early stage and evaluated their performance in this task. [18F]DMPY2 and [18F]DMPY3 were synthesized by direct radiofluorination of the bromo precursor, and radiochemical yields were ∼15–20%. Cell uptakes of [18F]DMPY2 and [18F]DMPY3 were >103-fold and 18-fold higher, respectively, in B16F10 (mouse melanoma) cells than in negative control cells. Biodistribution studies revealed strong tumor uptake and retention of [18F]DMPY2 (24.8% injected dose per gram of tissue [ID/g] at 60 min) and [18F]DMPY3 (11.7%ID/g at 60 min) in B16F10 xenografts. MicroPET imaging of both agents demonstrated strong tumoral uptake/retention and rapid washout, resulting in excellent tumor-to-background contrast in B16F10 xenografts. In particular, [18F]DMPY2 clearly visualized almost all metastatic lesions in lung and lymph nodes, with excellent image quality. [18F]DMPY2 demonstrated a significantly higher tumor-to-liver ratio than [18F]fluorodeoxyglucose ([18F]FDG) and the previously reported benzamide tracers N-[2-(diethylamino)-ethyl]-5-[18F]fluoropicolinamide ([18F]P3BZA) and N-[2-(diethylamino)-ethyl]-4-[18F]fluorobenzamide ([18F]FBZA) in B16F10-bearing or SK-MEL-3 (human melanoma)-bearing mice. In conclusion, [18F]DMPY2 might have strong potential for the diagnosis of early stage primary and metastatic melanoma using positron emission tomography (PET).
Collapse
|
13
|
Melanin-based nanomaterials: The promising nanoplatforms for cancer diagnosis and therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102211. [PMID: 32320736 DOI: 10.1016/j.nano.2020.102211] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 01/16/2023]
Abstract
Melanin-based nanoplatforms are biocompatible nanomaterials with a variety of unique physicochemical properties such as strong photothermal conversion ability, excellent drug binding capacity, strong metal chelation capacity, high chemical reactivity and versatile adhesion ability. These innate talents not only make melanin-based nanoplatforms be an inborn theranostic nanoagent for photoacoustic imaging-guided photothermal therapy of cancers, but also enable them to be conveniently transferred into cancer-targeting drug delivery systems and multimodality imaging nanoprobes. Due to the intriguing properties, melanin-based nanoplatforms have attracted much attention in investigations of cancer diagnosis and therapy. This review provides an overview of recent research advances in applications of melanin-based nanoplatforms in the fields of cancer diagnosis and therapy including cancer photothermal therapy, anticancer drug delivery, cancer-specific multimodal imaging and theranostics, etc. The remaining challenges and prospects of melanin-based nanoplatforms in biomedical applications are discussed at the end of this review.
Collapse
|
14
|
Zhang X, Chen F, Turker MZ, Ma K, Zanzonico P, Gallazzi F, Shah MA, Prater AR, Wiesner U, Bradbury MS, McDevitt MR, Quinn TP. Targeted melanoma radiotherapy using ultrasmall 177Lu-labeled α-melanocyte stimulating hormone-functionalized core-shell silica nanoparticles. Biomaterials 2020; 241:119858. [PMID: 32120314 DOI: 10.1016/j.biomaterials.2020.119858] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/27/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
Abstract
Lutetium-177 (177Lu) radiolabeled ultrasmall (~6 nm dia.) fluorescent core-shell silica nanoparticles (Cornell prime dots or C' dots) were developed for improving efficacy of targeted radiotherapy in melanoma models. PEGylated C' dots were surface engineered to display 10-15 alpha melanocyte stimulating hormone (αMSH) cyclic peptide analogs for targeting the melanocortin-1 receptor (MC1-R) over-expressed on melanoma tumor cells. The 177Lu-DOTA-αMSH-PEG-C' dot product was radiochemically stable, biologically active, and exhibited high affinity cellular binding properties and internalization. Selective tumor uptake and favorable biodistribution properties were also demonstrated, in addition to bulk renal clearance, in syngeneic B16F10 and human M21 xenografted models. Prolonged survival was observed in the treated cohorts relative to controls. Dosimetric analysis showed no excessively high absorbed dose among normal organs. Correlative histopathology of ex vivo treated tumor specimens revealed expected necrotic changes; no acute pathologic findings were noted in the liver or kidneys. Collectively, these results demonstrated that 177Lu-DOTA-αMSH-PEG-C' dot targeted melanoma therapy overcame the unfavorable biological properties and dose-limiting toxicities associated with existing mono-molecular treatments. The unique and tunable surface chemistries of this targeted ultrasmall radiotherapeutic, coupled with its favorable pharmacokinetic properties, substantially improved treatment efficacy and demonstrated a clear survival benefit in melanoma models, which supports its further clinical translation.
Collapse
Affiliation(s)
- Xiuli Zhang
- Harry S. Truman Veterans' Hospital, 800 Hospital Dr., Columbia, MO 65201, United States; Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States
| | - Feng Chen
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, United States
| | - Melik Z Turker
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, United States
| | - Kai Ma
- Elucida Oncology, New York, NY 10016, United States
| | - Pat Zanzonico
- Department of Medical Physics, Sloan Kettering Institute for Cancer Research, New York, NY 10065, United States
| | - Fabio Gallazzi
- Department of Chemistry and Research Core Facilities, University of Missouri, Columbia, MO 65211, United States
| | - Manankumar A Shah
- Harry S. Truman Veterans' Hospital, 800 Hospital Dr., Columbia, MO 65201, United States; Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States
| | - Austin R Prater
- Harry S. Truman Veterans' Hospital, 800 Hospital Dr., Columbia, MO 65201, United States; Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States
| | - Ulrich Wiesner
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, United States
| | - Michelle S Bradbury
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, United States; Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, United States
| | - Michael R McDevitt
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, United States
| | - Thomas P Quinn
- Harry S. Truman Veterans' Hospital, 800 Hospital Dr., Columbia, MO 65201, United States; Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States.
| |
Collapse
|
15
|
18F-Labeled Cyclized α-Melanocyte-Stimulating Hormone Derivatives for Imaging Human Melanoma Xenograft with Positron Emission Tomography. Sci Rep 2019; 9:13575. [PMID: 31537869 PMCID: PMC6753210 DOI: 10.1038/s41598-019-50014-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022] Open
Abstract
Since metastatic melanoma is deadly, early diagnosis thereof is crucial for managing the disease. We recently developed α-melanocyte-stimulating hormone (αMSH) derivatives, [68Ga]Ga-CCZ01048 and [18F]CCZ01064, that target the melanocortin 1 receptor (MC1R) for mouse melanoma imaging. In this study, we aim to evaluate [18F]CCZ01064 as well as a novel dual-ammoniomethyl-trifluoroborate (AmBF3) derivative, [18F]CCZ01096, for targeting human melanoma xenograft using μPET imaging. The peptides were synthesized on solid phase using Fmoc chemistry. Radiolabeling was achieved in a one-step 18F-19F isotope-exchange reaction. μPET imaging and biodistribution studies were performed in NSG mice bearing SK-MEL-1 melanoma xenografts. The MC1R density on the SK-MEL-1 cell line was determined to be 972 ± 154 receptors/cell (n = 4) via saturation assays. Using [18F]CCZ01064, moderate tumor uptake (3.05 ± 0.47%ID/g) and image contrast were observed at 2 h post-injection. Molar activity was determined to play a key role. CCZ01096 with two AmBF3 motifs showed comparable sub-nanomolar binding affinity to MC1R and much higher molar activity. This resulted in improved tumor uptake (6.46 ± 1.42%ID/g) and image contrast (tumor-to-blood and tumor-to-muscle ratios were 30.6 ± 5.7 and 85.7 ± 11.3, respectively) at 2 h post-injection. [18F]CCZ01096 represents a promising αMSH-based μPET imaging agent for human melanoma and warrants further investigation for potential clinical translation.
Collapse
|
16
|
Wang W, Jing T, Xia X, Tang L, Huang Z, Liu F, Wang Z, Ran H, Li M, Xia J. Melanin-loaded biocompatible photosensitive nanoparticles for controlled drug release in combined photothermal-chemotherapy guided by photoacoustic/ultrasound dual-modality imaging. Biomater Sci 2019; 7:4060-4074. [PMID: 31475710 DOI: 10.1039/c9bm01052a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Combined photothermal-chemotherapy guided by multimodal imaging is a promising strategy for cancer diagnosis and treatment. Multifunctional nanoparticles, such as those comprising organic and inorganic compounds, have been extensively investigated for combined photothermal-chemotherapy; however, their application is still limited by their potential long-term toxicity and lack of contrast properties. To solve these problems, in this study, a new type of multifunctional nanoparticle for combined photothermal-chemotherapy guided by dual-modality imaging was prepared with endogenous melanin by multistep emulsification to enhance tumor ablation. The nanoparticles were coated with poly(lactide-co-glycolic acid) (PLGA) and loaded with paclitaxel (PTX), encapsulated melanin and perfluoropentane (PFP). The materials in the nanoparticles were endogenous, ensuring high stability, biocompatibility, and biosafety. Nanoparticles irradiated with a laser, which induced their phase transformation into microbubbles, exhibited high photothermal conversion efficiency, thereby achieving photoacoustic (PA)/ultrasound (US) dual-modality imaging to determine tumor location, boundary, and size and to monitor drug distribution. Furthermore, optical droplet vaporization (ODV) of the nanoparticles could trigger the release of PTX; thus, these nanoparticles are a useful drug carrier. In vivo and in vitro experiments revealed that a strong synergistic antitumor effect was achieved by combining the photothermal properties of the nanoparticles with a chemotherapy drug. Importantly, the cavitation, thermoelastic expansion, and sonoporation caused by the phase transformation of the nanoparticles could directly damage the tumors. These processes also promoted the release, penetration and absorption of the drug, further enhancing the effect of combined photothermal-chemotherapy on tumor suppression. Therefore, the multifunctional nanoparticles prepared in this study provide a new strategy of using endogenous materials for controlled near-infrared (NIR)-responsive drug release and combined photothermal-chemotherapy guided by multimodal imaging.
Collapse
Affiliation(s)
- Wenyuan Wang
- Department of Ultrasound, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| | - Ting Jing
- Department of Radiology, Hospital (t.c.m) Affiliated to Southwest Medical University, Luzhou 646000, PR China
| | - Xiaorong Xia
- Department of Ultrasound, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| | - Linmei Tang
- Department of Ultrasound, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| | - Zhiqiang Huang
- Department of Ultrasound, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| | - Fengqiu Liu
- Institute of Ultrasound Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing 400010, PR China
| | - Zhigang Wang
- Institute of Ultrasound Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing 400010, PR China
| | - Haitao Ran
- Institute of Ultrasound Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing 400010, PR China
| | - Mingxing Li
- Department of Ultrasound, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| | - Jizhu Xia
- Department of Ultrasound, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| |
Collapse
|
17
|
Pyo A, Kim HS, Kim HS, Yun M, Kim DY, Min JJ. N-(2-(Dimethylamino)Ethyl)-4- 18F-Fluorobenzamide: A Novel Molecular Probe for High-Contrast PET Imaging of Malignant Melanoma. J Nucl Med 2018; 60:924-929. [PMID: 30552204 DOI: 10.2967/jnumed.118.221416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 11/30/2018] [Indexed: 01/06/2023] Open
Abstract
Malignant melanoma is an aggressive and serious form of skin cancer, with prognosis and treatment outcome depending heavily on the clinical stage of the disease at the time of diagnosis. Here, we synthesized a novel 18F-labeled benzamide derivative to target melanoma and then evaluated its biologic characteristics in small-animal models. Methods: N-(2-(dimethylamino)ethyl)-4-18F-fluorobenzamide (18F-DMFB) was synthesized by reaction of N-succinimidyl 4-18F-fluorobenzoate with N,N-dimethylethylenediamine. The binding affinity of 18F-DMFB was measured in B16F10 (mouse melanoma) cells with or without l-tyrosine. Small-animal PET imaging with 18F-DMFB was performed on B16F10 xenograft and metastasis mouse models. Results: The overall non-decay-corrected radiochemical yield of 18F-DMFB was approximately 10%-15%. Uptake of 18F-DMFB was melanin-specific, as cellular uptake in B16F10 increased more than 18-fold in the presence of l-tyrosine. Biodistribution studies revealed that 18F-DMFB accumulated, and was retained, in B16F10 xenografts for 120 min (10, 30, 60, and 120 min: 9.24, 10.80, 13.0, and 10.59 percentage injected dose/g, respectively) after radiotracer injection. Liver uptake of 18F-DMFB decreased from 10 to 120 min and showed fast clearance (10, 30, 60, and 120 min: 11.19, 5.7, 2.47, and 0.4 percentage injected dose/g). Furthermore, 18F-DMFB allowed visualization of metastatic lesions immediately after injection and was retained in lesions for over 60 min, with a high tumor-to-background ratio. Conclusion: 18F-DMFB demonstrated a high melanin-targeting ability and tumor-specific tumor uptake in both primary and metastatic lesions in animal models bearing malignant melanoma. 18F-DMFB may be a potential PET imaging agent for melanoma.
Collapse
Affiliation(s)
- Ayoung Pyo
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Korea
| | - Hyeon Sik Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Korea
| | - Hyung Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Hwasun, Korea; and
| | - Misun Yun
- Microbiology and Functionality Research Group, Research and Development Division, World Institute of Kimchi, Gwangju, Korea
| | - Dong-Yeon Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Korea
| |
Collapse
|
18
|
Zhang C, Lin KS, Bénard F. Molecular Imaging and Radionuclide Therapy of Melanoma Targeting the Melanocortin 1 Receptor. Mol Imaging 2018; 16:1536012117737919. [PMID: 29182034 PMCID: PMC5714078 DOI: 10.1177/1536012117737919] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Melanoma is a deadly disease at late metastatic stage, and early diagnosis and accurate staging remain the key aspects for managing melanoma. The melanocortin 1 receptor (MC1 R) is overexpressed in primary and metastatic melanomas, and its endogenous ligand, the α-melanocyte-stimulating hormone (αMSH), has been extensively studied for the development of MC1 R-targeted molecular imaging and therapy of melanoma. Natural αMSH is not well suited for this purpose due to low stability in vivo. Unnatural amino acid substitutions substantially stabilized the peptide, while cyclization via lactam bridge and metal coordination further improved binding affinity and stability. In this study, we summarized the development and the in vitro and in vivo characteristics of the radiolabeled αMSH analogues, including 99mTc-, 111In-, 67 Ga-, or 125I-labeled αMSH analogues for imaging with single-photon emission computed tomography; 68Ga-, 64Cu-, or 18F-labeled αMSH analogues for imaging with positron emission tomography; and 188Re-, 177Lu-, 90Y-, or 212Pb-labeled αMSH analogues for radionuclide therapy. These radiolabeled αMSH analogues showed promising results with high tumor uptake and rapid normal tissue activity clearance in the preclinical model of B16F1 and B16F10 mouse melanomas. These results highlight the potential of using radiolabeled αMSH analogues in clinical applications for molecular imaging and radionuclide therapy of melanoma.
Collapse
Affiliation(s)
- Chengcheng Zhang
- 1 Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Kuo-Shyan Lin
- 1 Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada.,2 Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - François Bénard
- 1 Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada.,2 Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
19
|
Wei W, Ehlerding EB, Lan X, Luo Q, Cai W. PET and SPECT imaging of melanoma: the state of the art. Eur J Nucl Med Mol Imaging 2018; 45:132-150. [PMID: 29085965 PMCID: PMC5700861 DOI: 10.1007/s00259-017-3839-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
Melanoma represents the most aggressive form of skin cancer, and its incidence continues to rise worldwide. 18F-FDG PET imaging has transformed diagnostic nuclear medicine and has become an essential component in the management of melanoma, but still has its drawbacks. With the rapid growth in the field of nuclear medicine and molecular imaging, a variety of promising probes that enable early diagnosis and detection of melanoma have been developed. The substantial preclinical success of melanin- and peptide-based probes has recently resulted in the translation of several radiotracers to clinical settings for noninvasive imaging and treatment of melanoma in humans. In this review, we focus on the latest developments in radiolabeled molecular imaging probes for melanoma in preclinical and clinical settings, and discuss the challenges and opportunities for future development.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Road, Shanghai, 200233, China
- Department of Radiology, University of Wisconsin-Madison, Room 7137, 1111 Highland Avenue, Madison, WI, 53705-2275, USA
| | - Emily B Ehlerding
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China.
| | - Quanyong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Road, Shanghai, 200233, China.
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Room 7137, 1111 Highland Avenue, Madison, WI, 53705-2275, USA.
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, 53705, USA.
| |
Collapse
|
20
|
Xu X, Yuan L, Yin L, Jiang Y, Gai Y, Liu Q, Wang Y, Zhang Y, Lan X. Synthesis and Preclinical Evaluation of 18F-PEG 3-FPN for the Detection of Metastatic Pigmented Melanoma. Mol Pharm 2017; 14:3896-3905. [PMID: 29037039 DOI: 10.1021/acs.molpharmaceut.7b00607] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although 18F-5-fluoro-N-(2-[diethylamino]ethyl)picolinamide (18F-5-FPN) is considered a promising radiopharmaceutical for PET imaging of melanoma, it accumulates at high concentrations in the liver. The aim in this research was to optimize the structure of 18F-5-FPN with triethylene glycol to reduce liver uptake as well as improve pharmacokinetics, and to evaluate its performance in detection of melanoma liver and lung metastases. 18F-PEG3-FPN was successfully prepared with a high radiolabeling yield (44.68% ± 5.99%) and radiochemical purity (>99%). The uptake of 18F-PEG3-FPN by pigmented B16F10 melanoma cells was significantly higher than that by amelanotic melanoma A375 cells. The binding to B16F10 cells could be blocked by excess 19F-PEG3-FPN. On small animal PET images, B16F10 tumors, but not A375 tumors, were clearly delineated after 18F-PEG3-FPN injection. More importantly, 18F-PEG3-FPN uptake by liver (2.27 ± 0.45 and 1.74 ± 0.35% ID/g, at 1 and 2 h) was significantly lower than that of 18F-5-FPN, and the lesions in lung and liver could be clearly detected by 18F-PEG3-FPN PET imaging in mouse models of pulmonary or hepatic metastases. Overall, we successfully synthesized 18F-PEG3-FPN, which has higher labeling efficacy and better in vivo pharmacokinetics along with lower liver uptake compared to 18F-5-FPN. This suggests 18F-PEG3-FPN as a candidate for pigmented melanoma liver and lung metastasis detection.
Collapse
Affiliation(s)
- Xiaodong Xu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Lujie Yuan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Lianglan Yin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yaqun Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yichun Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| |
Collapse
|
21
|
Garg PK, Nazih R, Wu Y, Grinevich VP, Garg S. Selective targeting of melanoma using N-(2-diethylaminoethyl) 4-[ 18F]fluoroethoxy benzamide (4-[ 18F]FEBZA): a novel PET imaging probe. EJNMMI Res 2017; 7:61. [PMID: 28791653 PMCID: PMC5548701 DOI: 10.1186/s13550-017-0311-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/25/2017] [Indexed: 02/04/2023] Open
Abstract
Background The purpose of this study was to develop a positron emission tomography (PET) imaging probe that is easy to synthesize and selectively targets melanoma in vivo. Herein, we report the synthesis and preclinical evaluation of N-(2-diethylaminoethyl) 4-[18F]Fluoroethoxy benzamide (4-[18F]FEBZA). A one-step synthesis was developed to prepare 4-[18F]FEBZA in high radiochemical yields and specific activity. The binding affinity, the in vitro binding, and internalization studies were performed using B16F1 melanoma cell line. The biodistribution studies were performed in C57BL/6 normal mice, C57BL/6 mice bearing B16F1 melanoma tumor xenografts, and nu/nu athymic mice bearing HT-29 human adenocarcinoma tumor and C-32 amelanotic melanoma tumor xenografts. MicroPET studies were performed in mice bearing B16F1 and HT-29 tumor xenografts. Results 4-[18F]FEBZA was prepared in 53 ± 14% radiochemical yields and a specific activity of 8.7 ± 1.1 Ci/μmol. The overall synthesis time for 4-[18F]FEBZA was 54 ± 7 min. The in vitro binding to B16F1 cells was 60.03 ± 0.48% after 1 h incubation at 37 °C. The in vivo biodistribution studies show a rapid and high uptake of F-18 in B16F1 tumor with 8.66 ± 1.02%IA/g in this tumor at 1 h. In contrast, the uptake at 1 h in HT-29 colorectal adenocarcinoma and C-32 amelanotic melanoma tumors was significantly lower with 3.68 ± 0.47%IA/g and 3.91 ± 0.23%IA/g in HT-29 and C-32 tumors, respectively. On microPET images, the melanoma tumor was clearly visible by 10 min post-injection and the intensity in the tumor continued to increase with time. In contrast, the HT-29 tumor was not visible on the microPET scans. Conclusions A rapid and facile synthesis of 4-[18F]FEBZA is developed. This method offers a reliable production of 4-[18F]FEBZA in high radiochemical yields and specific activity. A high binding affinity to melanoma cells and high uptake in tumor was noted. The microPET scan clearly delineates the melanoma tumor by 10 min post-injection. The results from these preclinical studies support the potential of 4-[18F]FEBZA as an effective probe to image melanoma.
Collapse
Affiliation(s)
- Pradeep K Garg
- Wake Forest University Health Sciences, Winston Salem, NC, USA. .,Center for Molecular Imaging and Therapy, Biomedical Research Foundation, 1505 Kings Highway, Shreveport, LA, 71133, USA.
| | - Rachid Nazih
- Wake Forest University Health Sciences, Winston Salem, NC, USA.,Center for Molecular Imaging and Therapy, Biomedical Research Foundation, 1505 Kings Highway, Shreveport, LA, 71133, USA
| | - Yanjun Wu
- Center for Molecular Imaging and Therapy, Biomedical Research Foundation, 1505 Kings Highway, Shreveport, LA, 71133, USA
| | - Vladimir P Grinevich
- Current Address: Asinex Corporation, 10 N. Chestnut Street, St 104, Winston Salem, NC, 27101, USA
| | - Sudha Garg
- Wake Forest University Health Sciences, Winston Salem, NC, USA.,Center for Molecular Imaging and Therapy, Biomedical Research Foundation, 1505 Kings Highway, Shreveport, LA, 71133, USA
| |
Collapse
|
22
|
Wang Y, Li M, Zhang Y, Zhang F, Liu C, Song Y, Zhang Y, Lan X. Detection of melanoma metastases with PET—Comparison of 18 F-5-FPN with 18 F–FDG. Nucl Med Biol 2017; 50:33-38. [DOI: 10.1016/j.nucmedbio.2017.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 03/22/2017] [Accepted: 03/30/2017] [Indexed: 12/19/2022]
|
23
|
Qian CG, Chen YL, Feng PJ, Xiao XZ, Dong M, Yu JC, Hu QY, Shen QD, Gu Z. Conjugated polymer nanomaterials for theranostics. Acta Pharmacol Sin 2017; 38:764-781. [PMID: 28552910 PMCID: PMC5520193 DOI: 10.1038/aps.2017.42] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/02/2017] [Indexed: 02/07/2023]
Abstract
Conjugated polymer nanomaterials (CPNs), as optically and electronically active materials, hold promise for biomedical imaging and drug delivery applications. This review highlights the recent advances in the utilization of CPNs in theranostics. Specifically, CPN-based in vivo imaging techniques, including near-infrared (NIR) imaging, two-photon (TP) imaging, photoacoustic (PA) imaging, and multimodal (MM) imaging, are introduced. Then, CPN-based photodynamic therapy (PDT) and photothermal therapy (PTT) are surveyed. A variety of stimuli-responsive CPN systems for drug delivery are also summarized, and the promising trends and translational challenges are discussed.
Collapse
Affiliation(s)
- Cheng-gen Qian
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Yu-lei Chen
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Pei-jian Feng
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xuan-zhong Xiao
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Mei Dong
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ji-cheng Yu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Quan-yin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Qun-dong Shen
- Department of Polymer Science and Engineering and Key Laboratory of High Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
24
|
Trencsényi G, Dénes N, Nagy G, Kis A, Vida A, Farkas F, Szabó JP, Kovács T, Berényi E, Garai I, Bai P, Hunyadi J, Kertész I. Comparative preclinical evaluation of 68Ga-NODAGA and 68Ga-HBED-CC conjugated procainamide in melanoma imaging. J Pharm Biomed Anal 2017; 139:54-64. [PMID: 28273651 DOI: 10.1016/j.jpba.2017.02.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/23/2017] [Accepted: 02/26/2017] [Indexed: 10/20/2022]
Abstract
Malignant melanoma is the most aggressive form of skin cancer. The early detection of primary melanoma tumors and metastases using non-invasive PET imaging determines the outcome of this disease. Previous studies have shown that benzamide derivatives (e.g. procainamide) conjugated with PET radionuclides specifically bind to melanin pigment of melanoma tumors. 68Ga chelating agents can have high influence on physiological properties of 68Ga labeled bioactive molecules, as was experienced during the application of HBED-CC on PSMA ligand. The aim of this study was to assess this concept in the case of the melanin specific procaindamide (PCA) and to compare the melanin specificity of 68Ga-labeled PCA using HBED-CC and NODAGA chelators under in vitro and in vivo conditions. Procainamide (PCA) was conjugated with HBED-CC and NODAGA chelators and was labeled with Ga-68. The melanin specificity of 68Ga-HBED-CC-PCA and 68Ga-NODAGA-PCA was investigated in vitro and in vivo using amelanotic (MELUR and A375) and melanin containing (B16-F10) melanoma cell lines. Tumor-bearing mice were prepared by subcutaneous injection of B16-F10, MELUR and A375 melanoma cells into C57BL/6 and SCID mice. 21±2days after tumor cell inoculation and 90min after intravenous injection of the 68Ga-labelledlabeled radiopharmacons whole body PET/MRI scans were performed. 68Ga-NODAGA-PCA and 68Ga-HBED-CC-PCA were produced with excellent radiochemical purity (98%). In vitro experiments demonstrated that after 30 and 90min incubation time 68Ga-NODAGA-PCA uptake of B16-F10 cells was significantly (p≤0.01) higher than the 68Ga-HBED-CC-conjugated PCA accumulation in the same cell line. Furthermore, significant difference (p≤0.01 and 0.05) was found between the uptake of melanin negative and positive cell lines using 68Ga-NODAGA-PCA and 68Ga-HBED-CC-PCA. In vivo PET/MRI studies using tumor models revealed significantly (p≤0.01) higher 68Ga-NODAGA-PCA uptake (SUVmean: 0.46±0.05, SUVmax: 1.96±0.25,T/M ratio: 40.7±4.23) in B16-F10 tumors in contrast to 68Ga-HBED-CC-PCA where the SUVmean, SUVmax and T/M ratio were 0.13±0.01, 0.56±0.11 and 11.43±1.24, respectively. Melanin specific PCA conjugated with NODAGA chelator showed higher specific binding properties than conjugated with HBED-CC. The chemical properties of the bifunctional chelators used for 68Ga-labeling of PCA determine the biological behaviour of the probes. Due to the high specificity and sensitivity 68Ga-labeled PCA molecules are promising radiotracers in melanoma imaging.
Collapse
Affiliation(s)
- György Trencsényi
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary; Scanomed LTD, Debrecen, Hungary.
| | - Noémi Dénes
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Adrienn Kis
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - András Vida
- Department of Medical Chemisty, University of Debrecen, Debrecen, Hungary; MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Flóra Farkas
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit P Szabó
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemisty, University of Debrecen, Debrecen, Hungary; MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Ervin Berényi
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Péter Bai
- Department of Medical Chemisty, University of Debrecen, Debrecen, Hungary; MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary; Research Center for Molecular Medicine, University of Debrecen, Hungary
| | - János Hunyadi
- Department of Dermatology, University of Debrecen, Debrecen, Hungary
| | - István Kertész
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
25
|
Kertész I, Vida A, Nagy G, Emri M, Farkas A, Kis A, Angyal J, Dénes N, Szabó JP, Kovács T, Bai P, Trencsényi G. In Vivo Imaging of Experimental Melanoma Tumors using the Novel Radiotracer 68Ga-NODAGA-Procainamide (PCA). J Cancer 2017; 8:774-785. [PMID: 28382139 PMCID: PMC5381165 DOI: 10.7150/jca.17550] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/10/2016] [Indexed: 12/14/2022] Open
Abstract
Purpose: The most aggressive form of skin cancer is the malignant melanoma. Because of its high metastatic potential the early detection of primary melanoma tumors and metastases using non-invasive PET imaging determines the outcome of the disease. Previous studies have already shown that benzamide derivatives, such as procainamide (PCA) specifically bind to melanin pigment. The aim of this study was to synthesize and investigate the melanin specificity of the novel 68Ga-labeled NODAGA-PCA molecule in vitro and in vivo using PET techniques. Methods: Procainamide (PCA) was conjugated with NODAGA chelator and was labeled with Ga-68 (68Ga-NODAGA-PCA). The melanin specificity of 68Ga-NODAGA-PCA was tested in vitro, ex vivo and in vivo using melanotic B16-F10 and amelanotic Melur melanoma cell lines. By subcutaneous and intravenous injection of melanoma cells tumor-bearing mice were prepared, on which biodistribution studies and small animal PET/CT scans were performed for 68Ga-NODAGA-PCA and 18FDG tracers. Results: 68Ga-NODAGA-PCA was produced with high specific activity (14.9±3.9 GBq/µmol) and with excellent radiochemical purity (98%<), at all cases. In vitro experiments showed that 68Ga-NODAGA-PCA uptake of B16-F10 cells was significantly (p≤0.01) higher than Melur cells. Ex vivo biodistribution and in vivo PET/CT studies using subcutaneous and metastatic tumor models showed significantly (p≤0.01) higher 68Ga-NODAGA-PCA uptake in B16-F10 primary tumors and lung metastases in comparison with amelanotic Melur tumors. In experiments where 18FDG and 68Ga-NODAGA-PCA uptake of B16-F10 tumors was compared, we found that the tumor-to-muscle (T/M) and tumor-to-lung (T/L) ratios were significantly (p≤0.05 and p≤0.01) higher using 68Ga-NODAGA-PCA than the 18FDG accumulation. Conclusion: Our novel radiotracer 68Ga-NODAGA-PCA showed specific binding to the melanin producing experimental melanoma tumors. Therefore, 68Ga-NODAGA-PCA is a suitable diagnostic radiotracer for the detection of melanoma tumors and metastases in vivo.
Collapse
Affiliation(s)
- István Kertész
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - András Vida
- Department of Medical Chemisty, University of Debrecen, Debrecen, Hungary;; MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | | | - Miklós Emri
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - Antal Farkas
- Department of Urology, University of Debrecen, Debrecen, Hungary
| | - Adrienn Kis
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - János Angyal
- Department of Periodontology, University of Debrecen, Debrecen, Hungary
| | - Noémi Dénes
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit P Szabó
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemisty, University of Debrecen, Debrecen, Hungary;; MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemisty, University of Debrecen, Debrecen, Hungary;; MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary;; Research Center for Molecular Medicine, University of Debrecen, Hungary
| | - György Trencsényi
- Department of Medical Imaging, Nuclear Medicine, University of Debrecen, Debrecen, Hungary;; Scanomed LTD, Debrecen, Hungary
| |
Collapse
|
26
|
Dasargyri A, Kümin CD, Leroux JC. Targeting Nanocarriers with Anisamide: Fact or Artifact? ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1603451. [PMID: 27885719 DOI: 10.1002/adma.201603451] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/08/2016] [Indexed: 05/19/2023]
Abstract
Encapsulating chemotherapeutics in nanoparticles can reduce the side effects of intravenous administration and improve their antitumor efficacy. Additionally, surface decoration of the nanocarriers with tumor-targeting ligands may enhance their specificity for cancer cells overexpressing the corresponding ligand-binding counterpart. The focus here is on anisamide, a low-molecular-weight benzamide derivative used as a tumor-directing moiety in functionalized nanosystems, based on its alleged interaction with Sigma receptors. The scintigraphic agents that initially inspired the use of anisamide for tumor targeting are described, and the published anisamide-tethered nanocarrier formulations are reviewed, together with a critical overview of the ligand's tumor-targeting properties. Moreover, anisamide's putative but dubious cellular target, the Sigma-1 receptor, is discussed with regard to its subcellular localization and implications in cancer. Data from in vivo studies reveal that the effect of anisamide on the antitumor efficacy of the decorated nanosystems varies considerably among the published reports. Together with the evidence questioning the interaction of anisamide with the Sigma receptors, the variability of anisamide's effect on the tumor deposition and the antitumor efficacy of the decorated drug carriers calls into question the extent of the ligand's tumor-targeting effect. Further research is necessary to elucidate the ligand's utility in tumor targeting.
Collapse
Affiliation(s)
- Athanasia Dasargyri
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, 8093, Switzerland
| | - Carole D Kümin
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, 8093, Switzerland
| | - Jean-Christophe Leroux
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, 8093, Switzerland
| |
Collapse
|
27
|
Garg PK, Nazih R, Wu Y, Singh R, Garg S. 4- 11C-Methoxy N-(2-Diethylaminoethyl) Benzamide: A Novel Probe to Selectively Target Melanoma. J Nucl Med 2016; 58:827-832. [PMID: 27980051 DOI: 10.2967/jnumed.116.184564] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/07/2016] [Indexed: 01/25/2023] Open
Abstract
We report the synthesis and preclinical evaluation of a 11C-labeled probe to target melanoma using PET. Methods: The target compound 4-11C-methoxy N-(2-diethylaminoethyl) benzamide (4-11C-MBZA) was prepared via the 11C-methylation of 4-hydroxy N-(2-diethylaminoethyl) benzamide (4-HBZA). The in vitro binding was performed using B16F1 (melanoma cells), MCF-10A (breast epithelial cells), and MDA-MB 231 (breast cancer cells). The internalization studies were conducted using B16F1 cells. In vivo biodistribution and small-animal PET imaging were performed in mice bearing B16F1 melanoma tumor xenografts. Results: The target compound 4-11C-MBZA was prepared in 46% ± 7% radiochemical yields by reacting 11C-methyltriflate with 4-HBZA followed by high-performance liquid chromatography purification. The specific activity of this compound was 853 ± 29.6 GBq/μmol (23 ± 0.8 Ci/μmol). The binding of 4-11C-MBZA to B16F1, MCF-10A, and MDA-MB-231 cells was 6.41% ± 1.28%, 1.51% ± 0.17%, and 0.30% ± 0.17%, respectively. Internalization studies using B16F1 melanoma cells show 60.7% of the cell-bound activity was internalized. Results from biodistribution studies show a rapid and high uptake of radioactivity in the tumor, with uptake levels reaching 5.85 ± 0.79 and 8.13 ± 1.46 percentage injected dose per gram at 10 and 60 min, respectively. Low uptake in normal tissues in conjunction with high tumor uptake resulted in high tumor-to-tissue ratios. On small-animal PET images, the tumor was clearly delineated soon after 4-11C-MBZA injection and tumor uptake reached 4.2 percentage injected dose per gram by 20 min. These preclinical evaluations show a high propensity of 4-11C-MBZA toward melanoma tumor. Conclusion: We successfully developed 4-11C-MBZA as a PET imaging probe, displaying properties advantageous over those for its 18F analogs. These preclinical evaluation results demonstrate the clinical potential of this probe to selectively target melanoma.
Collapse
Affiliation(s)
- Pradeep K Garg
- Department of Radiology, Wake Forest University Medical Center, Winston Salem, North Carolina .,Biomedical Research Foundation, Shreveport, Louisiana; and
| | - Rachid Nazih
- Department of Radiology, Wake Forest University Medical Center, Winston Salem, North Carolina.,Biomedical Research Foundation, Shreveport, Louisiana; and
| | - Yanjun Wu
- Biomedical Research Foundation, Shreveport, Louisiana; and
| | - Ravi Singh
- Department of Radiology, Wake Forest University Medical Center, Winston Salem, North Carolina.,Department of Cancer Biology, Wake Forest University Medical Center, Winston Salem, North Carolina
| | - Sudha Garg
- Department of Radiology, Wake Forest University Medical Center, Winston Salem, North Carolina .,Biomedical Research Foundation, Shreveport, Louisiana; and
| |
Collapse
|
28
|
Chang CC, Chang CH, Lo YH, Lin MH, Shen CC, Liu RS, Wang HE, Chen CL. Preparation and characterization of a novel Al(18)F-NOTA-BZA conjugate for melanin-targeted imaging of malignant melanoma. Bioorg Med Chem Lett 2016; 26:4133-9. [PMID: 27445169 DOI: 10.1016/j.bmcl.2016.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 11/16/2022]
Abstract
Melanin is an attractive target for the diagnosis and treatment of malignant melanoma. Previous studies have demonstrated the specific binding ability of benzamide moiety to melanin. In this study, we developed a novel (18)F-labeled NOTA-benzamide conjugate, Al(18)F-NOTA-BZA, which can be synthesized in 30min with a radiochemical yield of 20-35% and a radiochemical purity of >95%. Al(18)F-NOTA-BZA is highly hydrophilic (logP=-1.96) and shows good in vitro stability. Intravenous administration of Al(18)F-NOTA-BZA in two melanoma-bearing mouse models revealed highly specific uptake in B16F0 melanotic melanoma (6.67±0.91 and 1.50±0.26%ID/g at 15 and 120min p.i., respectively), but not in A375 amelanotic melanoma (0.87±0.21 and 0.24±0.09%ID/g at 15 and 120min p.i., respectively). The clearance from most normal tissues was fast. A microPET scan of Al(18)F-NOTA-BZA-injected mice also displayed high-contrast tumor images as compared with normal organs. Owing to the favorable in vivo distribution of Al(18)F-NOTA-BZA after intravenous administration, the estimated absorption dose was low in all normal organs and tissues. The melanin-specific binding ability, sustained tumor retention, fast normal tissues clearance and thelow projected human dosimetry supported that Al(18)F-NOTA-BZA is a very promising melanin-specific PET probe for melanin-positive melanoma.
Collapse
Affiliation(s)
- Chih-Chao Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, Pei-tou, Taipei 11221, Taiwan
| | - Chih-Hsien Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, Pei-tou, Taipei 11221, Taiwan; Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan, Taiwan
| | - Yi-Hsuan Lo
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, Pei-tou, Taipei 11221, Taiwan
| | - Ming-Hsien Lin
- Department of Nuclear Medicine, Taipei City Hospital, Zhongxiao Branch, Taipei, Taiwan
| | - Chih-Chieh Shen
- Department of Nuclear Medicine, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Ren-Shyan Liu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, Pei-tou, Taipei 11221, Taiwan; Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan; National PET/Cyclotron Center and Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Ell Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, Pei-tou, Taipei 11221, Taiwan.
| | - Chuan-Lin Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, Pei-tou, Taipei 11221, Taiwan.
| |
Collapse
|
29
|
Chang CC, Chang CH, Shen CC, Chen CL, Liu RS, Lin MH, Wang HE. Synthesis and characterization of a novel radioiodinated phenylacetamide and its homolog as theranostic agents for malignant melanoma. Eur J Pharm Sci 2015; 81:201-9. [PMID: 26517961 DOI: 10.1016/j.ejps.2015.10.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/16/2015] [Accepted: 10/25/2015] [Indexed: 01/22/2023]
Abstract
Melanin is an attractive target for the diagnosis and treatment of malignant melanoma. This study reports the preparation and biological characterizations of N-(2-(diethylamino)ethyl)-2-(3-(123/131)I-iodo-4- hydroxyphenyl)acetamide and N-(2-(diethylamino)ethyl)-3-(3-(123/131)I-iodo-4-hydroxyphenyl)propanamide (123/131)I-IHPA and 123/131I-IHPP) as novel melanin-specific theranostic agents. These two tracers were hydrophilic, exhibited good serum stability and high binding affinity to melanin. In vitro and in vivo studies revealed rapid, high and tenacious uptakes of both 131I-IHPA and 131I-IHPP in melanotic B16F0 cell line and in C57BL/6 mice bearing B16F0 melanoma, but not in amelanonic A375 cell line and tumors. Small-animal SPECT imaging also clearly delineate B16F0 melanoma since 1 h postinjection of 123I-IHPA and 123I-IHPP in tumor-bearing mice. Owing to the favorable biodistribution of 131I-IHPA and 131I-IHPP after intravenous administration, the estimated absorption dose was low in most normal organs and relatively high in melanotic tumor. The melanin-specific binding ability, sustained tumor retention, fast normal tissues clearance and acceptable projected human dosimetry supported that these two tracers are promising theranostic agents for melanin-positive melanoma.
Collapse
Affiliation(s)
- Chih-Chao Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Hsien Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; Isotope Application Division, Institute of Nuclear Energy Research, Taoyuan, Taiwan
| | - Chih-Chieh Shen
- Department of Nuclear Medicine, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Chuan-Lin Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ren-Shyan Liu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan; National PET/Cyclotron Center and Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Hsien Lin
- Department of Nuclear Medicine, Taipei City Hospital, Zhongxiao Branch, Taipei, Taiwan.
| | - Hsin-Ell Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
30
|
Zhang R, Fan Q, Yang M, Cheng K, Lu X, Zhang L, Huang W, Cheng Z. Engineering Melanin Nanoparticles as an Efficient Drug-Delivery System for Imaging-Guided Chemotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015. [PMID: 26222210 DOI: 10.1002/adma.201502201] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
In order to promote imaging-guided chemotherapy for preclinical and clinical applications, endogenous nanosystems with both contrast and drug-delivery properties are highly desired. Here, the simple use of melanin is first reported, and this biopolymer with good biocompatibility and biodegradability, binding ability to drugs and ions, and intrinsic photoacoustic properties, can serve as an efficient endogenous nanosystem for imaging-guided tumor chemotherapy in living mice.
Collapse
Affiliation(s)
- Ruiping Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, Stanford, CA, 94305-5484, USA
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Min Yang
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, Stanford, CA, 94305-5484, USA
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, China
| | - Kai Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, Stanford, CA, 94305-5484, USA
| | - Xiaomei Lu
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Lei Zhang
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Wei Huang
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, Stanford, CA, 94305-5484, USA
| |
Collapse
|
31
|
Feng H, Xia X, Li C, Song Y, Qin C, Liu Q, Zhang Y, Lan X. Imaging malignant melanoma with (18)F-5-FPN. Eur J Nucl Med Mol Imaging 2015; 43:113-122. [PMID: 26260649 DOI: 10.1007/s00259-015-3134-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/07/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE Radiolabelled benzamides are attractive candidates for targeting melanoma because they bind to melanin and exhibit high tumour uptake and retention. (18)F-5-Fluoro-N-(2-[diethylamino]ethyl)picolinamide ((18)F-5-FPN), a benzamide analogue, was prepared and its pharmacokinetics and binding affinity evaluated both in vitro and in vivo to assess its clinical potential in the diagnosis and staging of melanoma. METHODS (18)F-5-FPN was prepared and purified. Its binding specificity was measured in vitro in two different melanoma cell lines, one pigmented (B16F10 cells) and one nonpigmented (A375m cells), and in vivo in mice xenografted with the same cell lines. Dynamic and static PET images using (18)F-5-FPN were obtained in the tumour-bearing mice, and the static images were also compared with those acquired with (18)F-FDG. PET imaging with (18)F-5-FPN was also performed in B16F10 tumour-bearing mice with lung metastases. RESULTS (18)F-5-FPN was successfully prepared with radiochemical yields of 5 - 10 %. Binding of (18)F-5-FPN to B16F10 cells was much higher than to A375m cells. On dynamic PET imaging B16F10 tumours were visible about 1 min after injection of the tracer, and the uptake gradually increased over time. (18)F-5-FPN was rapidly excreted via the kidneys. B16F10 tumours were clearly visible on static images acquired 1 and 2 h after injection, with high uptake values of 24.34 ± 6.32 %ID/g and 16.63 ± 5.41 %ID/g, respectively, in the biodistribution study (five mice). However, there was no visible uptake by A375m tumours. (18)F-5-FPN and (18)F-FDG PET imaging were compared in B16F10 tumour xenografts, and the tumour-to-background ratio of (18)F-5-FPN was ten times higher than that of (18)F-FDG (35.22 ± 7.02 vs. 3.29 ± 0.53, five mice). (18)F-5-FPN PET imaging also detected simulated lung metastases measuring 1 - 2 mm. CONCLUSION (18)F-5-FPN specifically targeted melanin in vitro and in vivo with high retention and affinity and favourable pharmacokinetics. (18)F-5-FPN may be an ideal molecular probe for melanoma diagnosis and staging.
Collapse
Affiliation(s)
- Hongyan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China
| | - Chongjiao Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China
| | - Yiling Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China
| | - Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China.
| |
Collapse
|
32
|
Fan Q, Cheng K, Hu X, Ma X, Zhang R, Yang M, Lu X, Xing L, Huang W, Gambhir SS, Cheng Z. Transferring biomarker into molecular probe: melanin nanoparticle as a naturally active platform for multimodality imaging. J Am Chem Soc 2014; 136:15185-94. [PMID: 25292385 PMCID: PMC4227813 DOI: 10.1021/ja505412p] [Citation(s) in RCA: 293] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Indexed: 12/12/2022]
Abstract
Developing multifunctional and easily prepared nanoplatforms with integrated different modalities is highly challenging for molecular imaging. Here, we report the successful transfer of an important molecular target, melanin, into a novel multimodality imaging nanoplatform. Melanin is abundantly expressed in melanotic melanomas and thus has been actively studied as a target for melanoma imaging. In our work, the multifunctional biopolymer nanoplatform based on ultrasmall (<10 nm) water-soluble melanin nanoparticle (MNP) was developed and showed unique photoacoustic property and natural binding ability with metal ions (for example, (64)Cu(2+), Fe(3+)). Therefore, MNP can serve not only as a photoacoustic contrast agent, but also as a nanoplatform for positron emission tomography (PET) and magnetic resonance imaging (MRI). Traditional passive nanoplatforms require complicated and time-consuming processes for prebuilding reporting moieties or chemical modifications using active groups to integrate different contrast properties into one entity. In comparison, utilizing functional biomarker melanin can greatly simplify the building process. We further conjugated αvβ3 integrins, cyclic c(RGDfC) peptide, to MNPs to allow for U87MG tumor accumulation due to its targeting property combined with the enhanced permeability and retention (EPR) effect. The multimodal properties of MNPs demonstrate the high potential of endogenous materials with multifunctions as nanoplatforms for molecular theranostics and clinical translation.
Collapse
Affiliation(s)
- Quli Fan
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
- Key
Laboratory for Organic Electronics & Information Displays and
Institute of Advanced Materials, Nanjing
University of Posts & Telecommunications, Nanjing 210046, China
| | - Kai Cheng
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Xiang Hu
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Xiaowei Ma
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Ruiping Zhang
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Min Yang
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Xiaomei Lu
- Key
Laboratory for Organic Electronics & Information Displays and
Institute of Advanced Materials, Nanjing
University of Posts & Telecommunications, Nanjing 210046, China
| | - Lei Xing
- Department
of Radiation Oncology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Wei Huang
- Key
Laboratory for Organic Electronics & Information Displays and
Institute of Advanced Materials, Nanjing
University of Posts & Telecommunications, Nanjing 210046, China
| | - Sanjiv Sam Gambhir
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Zhen Cheng
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| |
Collapse
|
33
|
Beaino W, Anderson CJ. PET imaging of very late antigen-4 in melanoma: comparison of 68Ga- and 64Cu-labeled NODAGA and CB-TE1A1P-LLP2A conjugates. J Nucl Med 2014; 55:1856-63. [PMID: 25256059 DOI: 10.2967/jnumed.114.144881] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Melanoma is a malignant tumor derived from epidermal melanocytes, and it is known for its aggressiveness, therapeutic resistance, and predisposition for late metastasis. Very late antigen-4 (VLA-4; also called integrin α4β1) is a transmembrane noncovalent heterodimer overexpressed in melanoma tumors that plays an important role in tumor growth, angiogenesis, and metastasis by promoting adhesion and migration of cancer cells. In this study, we evaluated 2 conjugates of a high-affinity VLA-4 peptidomimetic ligand, LLP2A, for PET/CT imaging in a subcutaneous and metastatic melanoma tumor. METHODS LLP2A was conjugated to 1,4,8,11-tetraazacyclotetradecane-1-(methane phosphonic acid)-8-(methane carboxylic acid) (CB-TE1A1P) and 2-(4,7-bis(carboxymethyl)-1,4,7-triazonan-1-yl)pentanedioic acid (NODAGA) chelators for (68)Ga and (64)Cu labeling. The conjugates were synthesized by solid-phase peptide synthesis, purified by reversed-phase high-performance liquid chromatography, and verified by liquid chromatography mass spectrometry. Saturation and competitive binding assays with B16F10 melanoma cells determined the affinity of the compounds for VLA-4. The biodistributions of the LLP2A conjugates were evaluated in murine B16F10 subcutaneous tumor-bearing C57BL/6 mice. Melanoma metastasis was induced by intracardiac injection of B16F10 cells. PET/CT imaging was performed at 2, 4, and 24 h after injection for the (64)Cu tracers and 1 h after injection for the (68)Ga tracer. RESULTS (64)Cu-labeled CB-TE1A1P-PEG4-LLP2A and NODAGA-PEG4-LLP2A showed high affinity to VLA-4, with a comparable dissociation constant (0.28 vs. 0.23 nM) and receptor concentration (296 vs. 243 fmol/mg). The tumor uptake at 2 h after injection was comparable for the 2 probes, but (64)Cu-CB-TE1A1P-PEG4-LLP2A trended toward higher uptake than (64)Cu-NODAGA-PEG4-LLP2A (16.9 ± 2.2 vs. 13.4 ± 1.7 percentage injected dose per gram, P = 0.07). Tumor-to-muscle and tumor-to-blood ratios from biodistribution and PET/CT images were significantly higher for (64)Cu-CB-TE1A1P-PEG4-LLP2A than (64)Cu-NODAGA-PEG4-LLP2A (all P values < 0.05). PET/CT imaging of metastatic melanoma with (68)Ga-NODAGA-PEG4-LLP2A and (64)Cu-NODAGA-PEG4-LLP2A showed high uptake of the probes at the site of metastasis, correlating with the bioluminescence imaging of the tumor. CONCLUSION These data demonstrate that (64)Cu-labeled CB-TE1A1P/NODAGA LLP2A conjugates and (68)Ga-labeled NODAGA-LLP2A are excellent imaging agents for melanoma and potentially other VLA-4-positive tumors. (64)Cu-CB-TE1A1P-PEG4-LLP2A had the most optimal tumor-to-nontarget tissue ratios for translation into humans as a PET imaging agent for melanoma.
Collapse
Affiliation(s)
- Wissam Beaino
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Carolyn J Anderson
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania; and Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
34
|
Synthesis and preclinical characterization of [18F]FPBZA: a novel PET probe for melanoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:912498. [PMID: 25254219 PMCID: PMC4165567 DOI: 10.1155/2014/912498] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 08/03/2014] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Benzamide can specifically bind to melanoma cells. A 18F-labeled benzamide derivative, [18F]N-(2-diethylaminoethyl)-4-[2-(2-(2-fluoroethoxy) ethoxy)ethoxy]benzamide ([18F]FPBZA), was developed as a promising PET probe for primary and metastatic melanoma. METHODS [18F]FPBZA was synthesized via a one-step radiofluorination in this study. The specific uptake of [18F]FPBZA was studied in B16F0 melanoma cells, A375 amelanotic melanoma cells, and NB-DNJ-pretreated B16F0 melanoma cells. The biological characterization of [18F]FPBZA was performed on mice bearing B16F0 melanoma, A375 amelanotic melanoma, or inflammation lesion. RESULTS [18F]FPBZA can be prepared efficiently with a yield of 40-50%. The uptake of [18F]FPBZA by B16F0 melanoma cells was significantly higher than those by A375 tumor cells and NB-DNJ-pretreated B16F0 melanoma cells. B16F0 melanoma displayed prominent uptake of [18F]FPBZA at 2 h (7.81±0.82%ID/g), compared with A375 tumor and inflammation lesion (3.00±0.71 and 1.67±0.56%ID/g, resp.). [18F]FPBZA microPET scan clearly delineated B16F0 melanoma but not A375 tumor and inflammation lesion. In mice bearing pulmonary metastases, the lung radioactivity reached 4.77±0.36%ID/g at 2 h (versus 1.16±0.23%ID/g in normal mice). CONCLUSIONS Our results suggested that [18F]FPBZA PET would provide a promising and specific approach for the detection of primary and metastatic melanoma lesions.
Collapse
|
35
|
Gilardi L, Grana CM, Paganelli G. Evaluation of response to immunotherapy: new challenges and opportunities for PET imaging. Eur J Nucl Med Mol Imaging 2014; 41:2090-2. [PMID: 25012872 DOI: 10.1007/s00259-014-2848-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 06/24/2014] [Indexed: 01/13/2023]
Affiliation(s)
- Laura Gilardi
- Division of Nuclear Medicine, European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy,
| | | | | |
Collapse
|
36
|
Qin C, Cheng K, Chen K, Hu X, Liu Y, Lan X, Zhang Y, Liu H, Xu Y, Bu L, Su X, Zhu X, Meng S, Cheng Z. Tyrosinase as a multifunctional reporter gene for Photoacoustic/MRI/PET triple modality molecular imaging. Sci Rep 2014; 3:1490. [PMID: 23508226 PMCID: PMC3603217 DOI: 10.1038/srep01490] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/25/2013] [Indexed: 01/15/2023] Open
Abstract
Development of reporter genes for multimodality molecular imaging is highly important. In contrast to the conventional strategies which have focused on fusing several reporter genes together to serve as multimodal reporters, human tyrosinase (TYR)--the key enzyme in melanin production--was evaluated in this study as a stand-alone reporter gene for in vitro and in vivo photoacoustic imaging (PAI), magnetic resonance imaging (MRI) and positron emission tomography (PET). Human breast cancer cells MCF-7 transfected with a plasmid that encodes TYR (named as MCF-7-TYR) and non-transfected MCF-7 cells were used as positive and negative controls, respectively. Melanin targeted N-(2-(diethylamino)ethyl)-18F-5-fluoropicolinamide was used as a PET reporter probe. In vivo PAI/MRI/PET imaging studies showed that MCF-7-TYR tumors achieved significant higher signals and tumor-to-background contrasts than those of MCF-7 tumor. Our study demonstrates that TYR gene can be utilized as a multifunctional reporter gene for PAI/MRI/PET both in vitro and in vivo.
Collapse
Affiliation(s)
- Chunxia Qin
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University, Stanford, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Bu L, Li R, Liu H, Feng W, Xiong X, Zhao H, Vollrath D, Shen B, Cheng Z. Intrastriatal transplantation of retinal pigment epithelial cells for the treatment of Parkinson disease: in vivo longitudinal molecular imaging with 18F-P3BZA PET/CT. Radiology 2014; 272:174-83. [PMID: 24758555 DOI: 10.1148/radiol.14132042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To evaluate the performance of N-[2-(diethylamino)ethyl]-(18)F-5-fluoropicolinamide ((18)F-P3BZA) for visualizing porcine retinal pigment epithelium (pRPE) cells transplanted in the striatum for the treatment of Parkinson disease and to monitor the long-term activity of implanted pRPE cells by means of (18)F-P3BZA positron emission tomography (PET)/computed tomography (CT) in vivo. MATERIALS AND METHODS Animal work was conducted in accordance with the administrative panel on laboratory animal care. In vitro cell uptake of (18)F-P3BZA was determined with incubation of melanotic pRPE or amelanotic ARPE-19 cells with (18)F-P3BZA. To visualize the implanted pRPE cells in vivo, normal rats (four per group) were injected with pRPE or ARPE-19 cells attached to gelatin microcarriers in the left striatum and with control gelatin microcarriers in the right striatum and followed up with small animal PET/CT. Longitudinal PET/CT scans were acquired in 12 rats up to 16 days after surgery. Postmortem analysis, which included autoradiography and hematoxylin-eosin, Fontana-Masson, and immunofluorescence staining, was performed. Data were compared with the Student t test, analysis of variance, and regression analysis. RESULTS (18)F-P3BZA accumulated in pRPE cells effectively (3.48% of the injected dose [ID] per gram of brain tissue ± 0.58 at 1 hour after injection of the probe at 2 days after surgery in vivo) but not in control ARPE-19 cells (P < .05). Longitudinal PET/CT scans revealed that the activity of implanted pRPE cells decreased over time, as evidenced by a reduction in (18)F-P3BZA uptake (3.39% ID/g ± 0.18, 2.49% ID/g ± 0.41, and 1.20% ID/g ± 0.13 at days 2, 9, and 16, respectively; P < .05). Postmortem analysis helped confirm the results of in vivo imaging. CONCLUSION (18)F-P3BZA PET/CT is a feasible technique for visualizing and detecting the activity of implanted RPE cells in vivo.
Collapse
Affiliation(s)
- Lihong Bu
- From the Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University, 1201 Welch Rd, Lucas Center, Room P095, Stanford, CA 94305-5484 (L.B., R.L., H.L., Z.C.); Molecular Imaging Center, Department of Radiology, The 4th Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China (L.B., R.L., B.S.); and Departments of Genetics (W.F., D.V.) and Neurosurgery (X.X., H.Z.), School of Medicine, Stanford University, Stanford, Calif
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Qin C, Liu H, Chen K, Hu X, Ma X, Lan X, Zhang Y, Cheng Z. Theranostics of malignant melanoma with 64CuCl2. J Nucl Med 2014; 55:812-7. [PMID: 24627435 DOI: 10.2967/jnumed.113.133850] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Human copper transporter 1 (CTR1) is overexpressed in a variety of cancers. This study aimed to evaluate the use of (64)CuCl2 as a theranostic agent for PET and radionuclide therapy of malignant melanoma. METHODS CTR1 expression levels were detected by Western blot analysis of a group of tumor cell lines. Two melanoma cell lines (B16F10 and A375M) that highly expressed CTR1 were then selected to study the uptake and efflux of (64)CuCl2. Mice bearing B16F10 or A375M tumors (n = 4 for each group) were subjected to 5 min of static whole-body PET scans at different time points after intravenous injection of (64)CuCl2. Dynamic scans were also obtained for B16F10 tumor-bearing mice. All mice were sacrificed at 72 h after injection of (64)CuCl2, and biodistribution studies were performed. Mice bearing B16F10 or A375M tumors were further subjected to (64)CuCl2 radionuclide therapy. Specifically, when the tumor size reached 0.5-0.8 cm in diameter, tumor-bearing mice were systemically administered (64)CuCl2 (74 MBq) or phosphate-buffered saline, and tumor sizes were monitored over the treatment period. RESULTS CTR1 was found to be overexpressed in the cancer cell lines tested at different levels, and high expression levels in melanoma cells and tissues were observed (melanotic B16F10 and amelanotic A375M). (64)CuCl2 displayed high and specific uptake in B16F10 and A375M cells. In vivo (64)CuCl2 PET imaging demonstrated that both B16F10 and A375M tumors were clearly visualized. Radionuclide treatment studies showed that the tumor growth in both the B16F10 and the A375M models under (64)CuCl2 treatment were much slower than that of the control group. CONCLUSION Both melanotic and amelanotic melanomas (B16F10 and A375M) tested were found to overexpress CTR1. The tumors can be successfully visualized by (64)CuCl2 PET and further treated by (64)CuCl2, highlighting the high potential of using (64)CuCl2 as a theranostic agent for the management of melanoma.
Collapse
Affiliation(s)
- Chunxia Qin
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University, Stanford, California; and
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Liu H, Liu S, Miao Z, Jiang H, Deng Z, Hong X, Cheng Z. A novel aliphatic 18F-labeled probe for PET imaging of melanoma. Mol Pharm 2013; 10:3384-91. [PMID: 23927458 DOI: 10.1021/mp400225s] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Radiofluorinated benzamide and nicotinamide analogues are promising molecular probes for the positron emission tomography (PET) imaging of melanoma. Compounds containing aromatic (benzene or pyridine) and N,N-diethylethylenediamine groups have been successfully used for development of melanin targeted PET and single-photon emission computed tomography (SPECT) imaging agents for melanoma. The objective of this study was to determine the feasibility of using aliphatic compounds as a molecular platform for the development of a new generation of PET probes for melanoma detection. An aliphatic N,N-diethylethylenediamine precursor was directly coupled to a radiofluorination synthon, p-nitrophenyl 2-(18)F-fluoropropionate ((18)F-NFP), to produce the probe N-(2-(diethylamino)ethyl)-2-(18)F-fluoropropanamide ((18)F-FPDA). The melanoma-targeting ability of (18)F-FPDA was further evaluated both in vitro and in vivo through cell uptake assays, biodistribution studies, and small animal PET imaging in C57BL/6 mice bearing B16F10 murine melanoma tumors. Beginning with the precursor (18)F-NFP, the total preparation time for (18)F-FPDA, including the final high-performance liquid chromatography purification step, was approximately 30 min, with a decay-corrected radiochemical yield of 79.8%. The melanin-targeting specificity of (18)F-FPDA was demonstrated by significantly different uptake rates in tyrosine-treated and untreated B16F10 cells in vitro. The tumor uptake of (18)F-FPDA in vivo reached 2.65 ± 0.48 %ID/g at 2 h postinjection (p.i.) in pigment-enriched B16F10 xenografts, whereas the tumor uptake of (18)F-FPDA was close to the background levels, with rates of only 0.37 ± 0.07 %ID/g at 2 h p.i. in the nonpigmented U87MG tumor mouse model. Furthermore, small animal PET imaging studies revealed that (18)F-FPDA specifically targeted the melanotic B16F10 tumor, yielding a tumor-to-muscle ratio of approximately 4:1 at 1 h p.i. and 7:1 at 2 h p.i. In summary, we report the development of a novel (18)F-labeled aliphatic compound for melanoma imaging that can be easily synthesized in high yields using the radiosynthon (18)F-NFP. The PET probe (18)F-FPDA exhibits high B16F10 tumor-targeting efficacy and favorable in vivo pharmacokinetics. Our study demonstrates that aliphatic compounds can be used as a new generation molecular platform for the development of novel melanoma targeting agents. Further evaluation and optimization of (18)F-FPDA for melanin targeted molecular imaging are therefore warranted.
Collapse
Affiliation(s)
- Hongguang Liu
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, and Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University , California, 94305-5344, United States
| | | | | | | | | | | | | |
Collapse
|
40
|
Karlsson O, Lindquist NG. Melanin affinity and its possible role in neurodegeneration. J Neural Transm (Vienna) 2013; 120:1623-30. [PMID: 23821370 DOI: 10.1007/s00702-013-1062-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/24/2013] [Indexed: 12/22/2022]
Abstract
Certain drugs with melanin affinity are known to have caused pigmentary lesions in the eye and skin. This was the basis for the hypothesis that compounds with melanin affinity may cause damage also in other melanin-bearing tissues such as the substantia nigra. The heterogeneity of compounds that binds to melanin is large. Toxins, drugs, and several other compounds have melanin affinity. Compounds showing the highest affinity are mainly organic amines and metal ions. The binding of toxicants to melanin probably protects the cells initially. However, the binding is normally, slowly reversible and melanin may accumulate the toxicant and gradually release it into the cytosol. Several studies indicate that neuromelanin may play a significant role both in the initiation and in the progression of neurodegeneration. MPTP/MPP(+) that has been causally linked with Parkinsonism has high affinity for neuromelanin, and the induced dopaminergic denervation correlates with the neuromelanin content in the cells. This shows that the toxicological implications of the accumulation of toxicants in pigmented neurons and its possible role in neurodegeneration should not be neglected. Extracellular neuromelanin has been reported to activate dendritic cells and microglia. An initial neuronal damage induced by a neurotoxicant that leaks neuromelanin from the cells may therefore lead to a vicious cycle of neuroinflammation and further neurodegeneration. Although there are many clues to the particular vulnerability of dopaminergic neurons of substantia nigra in Parkinson's disease, the critical factors are not known. Further studies to determine the importance of neuromelanin in neurodegeneration and Parkinson's disease are warranted.
Collapse
Affiliation(s)
- Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 751 24, Uppsala, Sweden,
| | | |
Collapse
|
41
|
Synthesis, radioiodination and in vivo screening of novel potent iodinated and fluorinated radiotracers as melanoma imaging and therapeutic probes. Eur J Med Chem 2013; 63:840-53. [DOI: 10.1016/j.ejmech.2012.11.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 11/14/2012] [Accepted: 11/17/2012] [Indexed: 11/18/2022]
|
42
|
Liu H, Liu S, Miao Z, Deng Z, Shen B, Hong X, Cheng Z. Development of 18F-labeled picolinamide probes for PET imaging of malignant melanoma. J Med Chem 2013; 56:895-901. [PMID: 23301672 DOI: 10.1021/jm301740k] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Melanoma is an aggressive skin cancer with worldwide increasing incidence. Development of positron emission tomography (PET) probes for early detection of melanoma is critical for improving the survival rate of melanoma patients. In this research, (18)F-picolinamide-based PET probes were prepared by direct radiofluorination of the bromopicolinamide precursors using no-carrier-added (18)F-fluoride. The resulting probes, (18)F-1, (18)F-2 and (18)F-3, were then evaluated in vivo by small animal PET imaging and biodistribution studies in C57BL/6 mice bearing B16F10 murine melanoma tumors. Noninvasive small animal PET studies demonstrated excellent tumor imaging contrasts for all probes, while (18)F-2 showed higher tumor to muscle ratios than (18)F-1 and (18)F-3. Furthermore, (18)F-2 demonstrated good in vivo stability as evidenced by the low bone uptake in biodistribution studies. Collectively, these findings suggest (18)F-2 as a highly promising PET probe for translation into clinical detection of melanoma.
Collapse
Affiliation(s)
- Hongguang Liu
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, Department of Radiology, Stanford University, California, 94305-5344, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Taylor SR, Roberts MP, Wyatt NA, Pham TQ, Stark D, Bourdier T, Roselt P, Katsifis A, Greguric I. Synthesis and Radiosynthesis of a Novel PET Fluorobenzyl Piperazine for Melanoma Tumour Imaging; [18F]MEL054. Aust J Chem 2013. [DOI: 10.1071/ch12489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
2-{2-[4-(4-[18F]-Fluorobenzyl)piperazin-1-yl]-2-oxoethyl}isoindolin-1-one ([18F]MEL054), is a new potent indolinone-based melanin binder designed to target melanotic tumours. [18F]MEL054 was prepared by an automated two-step radiosynthesis, comprising of the preparation of 4-[18F]fluorobenzaldehyde from 4-formyl-N,N,N-trimethylanilinium triflate, followed by reductive alkylation with 2-(2-oxo-2-piperazin-1-ylethyl)isoindolin-1-one. 4-[18F]Fluorobenzaldehyde was prepared on a GE TRACERlab FXFN module in 68 ± 8 % radiochemical yield (RCY, non-decay corrected), purified by a Sep-Pak Plus C18 cartridge and eluted into the reactor of an in-house modified Nuclear Interface [18F]FDG synthesis module for the subsequent reductive alkylation reaction. HPLC purification produced [18F]MEL054 in a collected RCY of 34 ± 9 % (non-decay corrected), the total preparation time (including Sep-Pak Plus C18 and HPLC purification) did not exceed 105 min. The radiochemical purity of [18F]MEL054 was greater than 99 % with a specific radioactivity of 71–119 GBq μmol–1 and [18F]MEL054 remained stable in saline solution (>98 %) after 3 h.
Collapse
|
44
|
An Endogenous Electron Spin Resonance (ESR) signal discriminates nevi from melanomas in human specimens: a step forward in its diagnostic application. PLoS One 2012; 7:e48849. [PMID: 23144997 PMCID: PMC3492252 DOI: 10.1371/journal.pone.0048849] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 10/01/2012] [Indexed: 02/03/2023] Open
Abstract
Given the specific melanin-associated paramagnetic features, the Electron Spin Resonance (ESR, called also Electron Paramagnetic Resonance, EPR) analysis has been proposed as a potential tool for non-invasive melanoma diagnosis. However, studies comparing human melanoma tissues to the most appropriate physiological counterpart (nevi) have not been performed, and ESR direct correlation with melanoma clinical features has never been investigated. ESR spectrum was obtained from melanoma and non-melanoma cell-cultures as well as mouse melanoma and non-melanoma tissues and an endogenous ESR signal (g = 2.005) was found in human melanoma cells and in primary melanoma tissues explanted from mice, while it was always absent in non-melanoma samples. These characteristics of the measured ESR signal strongly suggested its connection with melanin. Quantitative analyses were then performed on paraffin-embedded human melanoma and nevus sections, and validated on an independent larger validation set, for a total of 112 sections (52 melanomas, 60 nevi). The ESR signal was significantly higher in melanomas (p = 0.0002) and was significantly different between “Low Breslow’s and “High Breslow’s” depth melanomas (p<0.0001). A direct correlation between ESR signal and Breslow’s depth, expressed in millimetres, was found (R = 0.57; p<0.0001). The eu/pheomelanin ratio was found to be significantly different in melanomas “Low Breslow’s” vs melanomas “High Breslow’s” depth and in nevi vs melanomas “High Breslow’s depth”. Finally, ROC analysis using ESR data discriminated melanomas sections from nevi sections with up to 90% accuracy and p<0.0002. In the present study we report for the first time that ESR signal in human paraffin-embedded nevi is significantly lower than signal in human melanomas suggesting that spectrum variations may be related to qualitative melanin differences specifically occurring in melanoma cells. We therefore conclude that this ESR signal may represent a reliable marker for melanoma diagnosis in human histological sections.
Collapse
|
45
|
Rbah-Vidal L, Vidal A, Besse S, Cachin F, Bonnet M, Audin L, Askienazy S, Dollé F, Degoul F, Miot-Noirault E, Moins N, Auzeloux P, Chezal JM. Early detection and longitudinal monitoring of experimental primary and disseminated melanoma using [¹⁰F]ICF01006, a highly promising melanoma PET tracer. Eur J Nucl Med Mol Imaging 2012; 39:1449-1461. [PMID: 22707183 DOI: 10.1007/s00259-012-2168-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 05/22/2012] [Indexed: 01/10/2023]
Abstract
PURPOSE Here, we report a new and rapid radiosynthesis of (18)F-N-[2-(diethylamino)ethyl]-6-fluoro-pyridine-3-carboxamide ([(18)F]ICF01006), a molecule with a high specificity for melanotic tissue, and its evaluation in a murine model for early specific detection of pigmented primary and disseminated melanoma. METHODS [(18)F]ICF01006 was synthesized using a new one-step bromine-for-fluorine nucleophilic heteroaromatic substitution. Melanoma models were induced by subcutaneous (primary tumour) or intravenous (lung colonies) injection of B16BL6 melanoma cells in C57BL/6J mice. The relevance and sensitivity of positron emission tomography (PET) imaging using [(18)F]ICF01006 were evaluated at different stages of tumoural growth and compared to (18)F-fluorodeoxyglucose ([(18)F]FDG). RESULTS The fully automated radiosynthesis of [(18)F]ICF01006 led to a radiochemical yield of 61 % and a radiochemical purity >99 % (specific activity 70-80 GBq/μmol; total synthesis time 42 min). Tumours were visualized before they were palpable as early as 1 h post-injection with [(18)F]ICF01006 tumoural uptake of 1.64 ± 0.57, 3.40 ± 1.47 and 11.44 ± 2.67 percentage of injected dose per gram of tissue (%ID/g) at days 3, 5 and 14, respectively. [(18)F]ICF01006 PET imaging also allowed detection of melanoma pulmonary colonies from day 9 after tumour cell inoculation, with a lung radiotracer accumulation correlated with melanoma invasion. At day 21, radioactivity uptake in lungs reached a value of 5.23 ± 2.08 %ID/g (versus 0.41 ± 0.90 %ID/g in control mice). In the two models, comparison with [(18)F]FDG showed that both radiotracers were able to detect melanoma lesions, but [(18)F]ICF01006 was superior in terms of contrast and specificity. CONCLUSION Our promising results provide further preclinical data, reinforcing the excellent potential of [(18)F]ICF01006 PET imaging for early specific diagnosis and follow-up of melanin-positive disseminated melanoma.
Collapse
Affiliation(s)
- Latifa Rbah-Vidal
- Imagerie Moléculaire et Thérapie Vectorisée, Clermont Université, Université d'Auvergne, BP 10448, 63000 Clermont-Ferrand, France,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kim HJ, Kim DY, Park JH, Yang SD, Hur MG, Min JJ, Yu KH. Synthesis and evaluation of a novel 68Ga-labeled DOTA-benzamide derivative for malignant melanoma imaging. Bioorg Med Chem Lett 2012; 22:5288-92. [DOI: 10.1016/j.bmcl.2012.06.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 06/12/2012] [Accepted: 06/13/2012] [Indexed: 11/27/2022]
|
47
|
Synthesis and characterization of a (68)Ga-labeled N-(2-diethylaminoethyl)benzamide derivative as potential PET probe for malignant melanoma. Bioorg Med Chem 2012; 20:4915-20. [PMID: 22831797 DOI: 10.1016/j.bmc.2012.06.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/27/2012] [Accepted: 06/27/2012] [Indexed: 10/28/2022]
Abstract
Radiolabeled benzamides have been reported to be attractive agents for targeting malignant melanoma as they bind melanin and display high accumulation in melanoma cells. Herein, we report the synthesis and bioevaluation of a novel (68)Ga-labeled benzamide as a potential PET agent for malignant melanoma. The novel radiotracer was synthesized in good radiochemical yields (80% decay corrected yield) and high specific radioactivity (10 GBq/μmol). Cellular uptake of (68)Ga-SCN-NOTA-BZA was significantly higher in B16F10 cells (mouse melanoma) treated with L-tyrosine. Biodistribution and micro-PET studies of (68)Ga-SCN-NOTA-BZA in B16F10-bearing mice showed selective uptake into the tumor. The radiotracer was cleared via renal excretion without further metabolism. These results demonstrate that (68)Ga-SCN-NOTA-BZA is a potential PET probe for malignant melanoma.
Collapse
|
48
|
Abstract
Despite recognizing the devastating consequences of metastasis, we are not yet able to effectively treat cancer that has spread to vital organs. The inherent complexity of genomic alterations in late-stage cancers, coupled with numerous heterotypic interactions that occur between tumour and stromal cells, represent fundamental challenges in our quest to understand and control metastatic disease. The incorporation of genomic and other systems level approaches, as well as technological breakthroughs in imaging and animal modelling, have galvanized the effort to overcome gaps in our understanding of metastasis. Future research carries with it the potential to translate the wealth of new knowledge and conceptual advances into effective targeted therapies.
Collapse
Affiliation(s)
- Nilay Sethi
- Department of Molecular Biology, Washington Road, LTL 255, Princeton University, Princeton, New Jersey 08544, USA
| | | |
Collapse
|
49
|
Maisonial A, Kuhnast B, Papon J, Boisgard R, Bayle M, Vidal A, Auzeloux P, Rbah L, Bonnet-Duquennoy M, Miot-Noirault E, Galmier MJ, Borel M, Askienazy S, Dollé F, Tavitian B, Madelmont JC, Moins N, Chezal JM. Single photon emission computed tomography/positron emission tomography imaging and targeted radionuclide therapy of melanoma: new multimodal fluorinated and iodinated radiotracers. J Med Chem 2011; 54:2745-2766. [PMID: 21417462 DOI: 10.1021/jm101574q] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This study reports a series of 14 new iodinated and fluorinated compounds offering both early imaging ((123)I, (124)I, (18)F) and systemic treatment ((131)I) of melanoma potentialities. The biodistribution of each (125)I-labeled tracer was evaluated in a model of melanoma B16F0-bearing mice, using in vivo serial γ scintigraphic imaging. Among this series, [(125)I]56 emerged as the most promising compound in terms of specific tumoral uptake and in vivo kinetic profile. To validate our multimodality concept, the radiosynthesis of [(18)F]56 was then optimized and this radiotracer has been successfully investigated for in vivo PET imaging of melanoma in B16F0- and B16F10-bearing mouse model. The therapeutic efficacy of [(131)I]56 was then evaluated in mice bearing subcutaneous B16F0 melanoma, and a significant slow down in tumoral growth was demonstrated. These data support further development of 56 for PET imaging ((18)F, (124)I) and targeted radionuclide therapy ((131)I) of melanoma using a single chemical structure.
Collapse
Affiliation(s)
- Aurélie Maisonial
- Clermont Université, Université d'Auvergne, Imagerie Moléculaire et Thérapie Vectorisée, Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Greguric I, Taylor S, Pham T, Wyatt N, Jiang CD, Bourdier T, Loc'h C, Roselt P, Neels OC, Katsifis A. Radiosynthesis of a Novel PET Fluoronicotinamide for Melanoma Tumour PET Imaging; [18F]MEL050. Aust J Chem 2011. [DOI: 10.1071/ch11048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
[18F]6-Fluoro-N-[2-(diethylamino)ethyl]nicotinamide [18F]MEL050 is a novel nicotinamide-based radiotracer, designed to target random metastatic dissemination of melanoma tumours by targeting melanin. Preclinical studies suggest that [18F]MEL050 has an excellent potential to improve diagnosis and staging of melanoma. Here we report the radiochemical optimization conditions of [18F]MEL050 and its large scale automated synthesis using a GE FXFN automated radiosynthesis module for clinical, phase-1 investigation. [18F]MEL050 was prepared via a one-step synthesis using no-carrier added K[18F]F-Krytpofix® 222 (DMSO, 170°C, 5 min) followed by HPLC purification. Using 6-chloro-N-[2-(diethylamino)ethyl]nicotinamide as precursor, [18F]MEL050 was obtained in 40–46% radiochemical yield (non-decay corrected), in greater than 99.9% radiochemical purity and specific activity ranging from 240 to 325 GBq μmol–1. Total synthesis time including formulation was 40 min and [18F]MEL050 was stable (99.8%) in PBS for 6 h.
Collapse
|