1
|
Chambers C, Chitwood B, Smith CJ, Miao Y. Elevating theranostics: The emergence and promise of radiopharmaceutical cell-targeting heterodimers in human cancers. IRADIOLOGY 2024; 2:128-155. [PMID: 38708130 PMCID: PMC11067702 DOI: 10.1002/ird3.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/30/2024] [Indexed: 05/07/2024]
Abstract
Optimal therapeutic and diagnostic efficacy is essential for healthcare's global mission of advancing oncologic drug development. Accurate diagnosis and detection are crucial prerequisites for effective risk stratification and personalized patient care in clinical oncology. A paradigm shift is emerging with the promise of multi-receptor-targeting compounds. While existing detection and staging methods have demonstrated some success, the traditional approach of monotherapy is being reevaluated to enhance therapeutic effectiveness. Heterodimeric site-specific agents are a versatile solution by targeting two distinct biomarkers with a single theranostic agent. This review describes the innovation of dual-targeting compounds, examining their design strategies, therapeutic implications, and the promising path they present for addressing complex diseases.
Collapse
Affiliation(s)
- Claudia Chambers
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
- Research Division, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, USA
- Department of Chemistry, University of Missouri, Columbia, Missouri, USA
| | - Broc Chitwood
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
| | - Charles J. Smith
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
- Research Division, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, USA
- Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri, USA
| | - Yubin Miao
- Department of Radiology, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
2
|
Ni Q, Li X, Huang H, Ge Z. Decreased expression of SCARA5 predicts a poor prognosis in melanoma using bioinformatics analysis. Front Oncol 2023; 13:1015358. [PMID: 37035142 PMCID: PMC10079878 DOI: 10.3389/fonc.2023.1015358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 02/24/2023] [Indexed: 04/11/2023] Open
Abstract
Background It has been established that the scavenger receptor class A member 5 (SCARA5) functions as a tumor suppressor gene in various cancer types. To our knowledge, no comprehensive study has hitherto investigated the expression and function of SCARA5 in melanoma. This study aimed to determine the association between SCARA5 and melanoma. Methods Analysis of SCARA5 mRNA expression was performed using The Cancer Genome Atlas (TCGA) data sets. To evaluate the clinical significance of SCARA5, the clinical data of 93 patients with melanoma were collected. The role of SCARA5 expression in prognosis was also analyzed. In this study, survival was evaluated by Kaplan-Meier analysis and compared using the log-rank test. Univariate and multivariate Cox proportional hazard regression analyses were used to identify independent predictors. The Kyoto Encyclopedia of Genes and Genomes, Gene Ontology, and gene set enrichment analysis (GSEA) were used to perform gene set functional annotations. Protein-protein interaction (PPI) networks were constructed to illustrate gene-gene interactions. The Tumor IMmune Estimation Resource (TIMER) database was used to explore the association between SCARA5 and immune infiltration levels. Results The results showed that the SCARA5 mRNA expression in melanoma was significantly lower than in adjacent normal skin tissue (p < 0.001). Moreover, decreased expression of SCARA5 in melanoma correlated with the tumor, node, and metastasis (TNM) stage and recurrence (p < 0.05). The overall survival (OS) was significantly higher in melanoma with high SCARA5 expression compared with low SCARA5 expression (p < 0.001). During univariate analysis, SCARA5 expression, tumor (T) stage, node (N) stage, metastasis (M) stage, and recurrence correlated with OS (p < 0.05). Further multivariate Cox regression analysis showed that SCARA5 expression (p = 0.012) could be an independent prognostic factor for OS in cutaneous malignant melanoma. GSEA analysis showed that SCARA5 was significantly enriched in various pathways, such as response to developmental biology and response to antimicrobial peptides. Correlation analysis showed a positive correlation with CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells (p < 0.05), and a negative correlation with tumor purity (p < 0.05). Conclusion SCARA5 has significant potential as a prognostic biomarker and as a promising therapeutic target in melanoma. Furthermore, SCARA5 expression in melanoma is related to the level of immune infiltration.
Collapse
Affiliation(s)
- Qinggan Ni
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Burns and Plastic Surgery, Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, Yancheng, China
| | - Xia Li
- Department of General Medicine, Yancheng Third People’s Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China
| | - Hua Huang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zili Ge
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- *Correspondence: Zili Ge,
| |
Collapse
|
3
|
Development and Evaluation of a Peptide Heterodimeric Tracer Targeting CXCR4 and Integrin α vβ 3 for Pancreatic Cancer Imaging. Pharmaceutics 2022; 14:pharmaceutics14091791. [PMID: 36145541 PMCID: PMC9503769 DOI: 10.3390/pharmaceutics14091791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Nowadays, pancreatic cancer is still a formidable disease to diagnose. The CXC chemokine receptor 4 (CXCR4) and integrin αvβ3 play important roles in tumor development, progression, invasion, and metastasis, which are overexpressed in many types of human cancers. In this study, we developed a heterodimeric tracer 68Ga-yG5-RGD targeting both CXCR4 and integrin αvβ3, and evaluated its feasibility and utility in PET imaging of pancreatic cancer. The 68Ga-yG5-RGD could accumulate in CXCR4/integrin αvβ3 positive BxPC3 tumors in a high concentration and was much higher than that of 68Ga-yG5 (p < 0.001) and 68Ga-RGD (p < 0.001). No increased uptake of 68Ga-yG5-RGD was found in MX-1 tumors (CXCR4/integrin αvβ3, negative). In addition, the uptake of 68Ga-yG5-RGD in BxPC3 was significantly blocked by excess amounts of AMD3100 (an FDA-approved CXCR4 antagonist) and/or unlabeled RGD (p < 0.001), confirming its dual-receptor targeting properties. The ex vivo biodistribution and immunohistochemical results were consistent with the in vivo imaging results. The dual-receptor targeting strategy achieved improved tumor-targeting efficiency and prolonged tumor retention in BxPC3 tumors, suggesting 68Ga-yG5-RGD is a promising tracer for the noninvasive detection of tumors that express either CXCR4 or integrin αvβ3 or both, and therefore may have good prospects for clinical translation.
Collapse
|
4
|
Zhao H, Gao H, Luo C, Yang G, Zhao X, Gao S, Ma Q, Jia B, Shi J, Wang F. An Integrin-α vβ 6/α 5β 1-Bitargeted Probe for the SPECT Imaging of Pancreatic Adenocarcinoma in Preclinical and Primary Clinical Studies. Bioconjug Chem 2021; 32:1298-1305. [PMID: 34137602 DOI: 10.1021/acs.bioconjchem.1c00296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pancreatic adenocarcinoma (PA) is one of the deadliest human malignancies. However, early detection, prediction of surgical resectability, and prognosis of PA are challenging with current conventional imaging technologies in the clinic. Molecular imaging technologies combined with novel imaging probes could be useful for early detection and accurate staging of PA. Integrin αvβ6 and α5β1 are found to be overexpressed in PA. In this study, integrin αvβ6/α5β1-bitargeted probes 99mTc-HYNIC-isoDGR (99mTc-isoDGR) and 99mTc-HYNIC-PEG4-PisoDGR2 (99mTc-3PisoDGR2) were prepared and evaluated in the BxPC-3 human pancreatic tumor model. Both subcutaneous and in situ BxPC-3 tumors could be clearly visualized by 99mTc-isoDGR nanoScan SPECT/CT imaging with a high ratio of tumor to background. The blocking study with excess nonradioactive peptide showed a significantly reduced tumor uptake, which confirmed the specificity of 99mTc-isoDGR. Biodistribution results confirmed the imaging results. The dimer tracer 99mTc-3PisoDGR2 significantly enhanced tumor uptake compared with 99mTc-isoDGR, and the spontaneous PA lesion in the mouse model could be clearly visualized by 99mTc-3PisoDGR2. The primary clinical study also verified the ability of 99mTc-3PisoDGR2 for detection of PA. Therefore, SPECT/CT imaging using the integrin αvβ6/α5β1-bitargeted 99mTc-3PisoDGR2 provided a potential approach for the noninvasive detection of PA.
Collapse
Affiliation(s)
- Haitao Zhao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hannan Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Chuangwei Luo
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Guangjie Yang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xiaoyu Zhao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Shi Gao
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, China.,NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, Jilin 130021, China
| | - Qingjie Ma
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, China.,NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, Jilin 130021, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jiyun Shi
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.,Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
5
|
Cheng X, Hübner R, von Kiedrowski V, Fricker G, Schirrmacher R, Wängler C, Wängler B. Design, Synthesis, In Vitro and In Vivo Evaluation of Heterobivalent SiFA lin-Modified Peptidic Radioligands Targeting Both Integrin α vβ 3 and the MC1 Receptor-Suitable for the Specific Visualization of Melanomas? Pharmaceuticals (Basel) 2021; 14:ph14060547. [PMID: 34200477 PMCID: PMC8228600 DOI: 10.3390/ph14060547] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Combining two peptides addressing two different receptors to a heterobivalent peptidic ligand (HBPL) is thought to enable an improved tumor-targeting sensitivity and thus tumor visualization, compared to monovalent peptide ligands. In the case of melanoma, the Melanocortin-1 receptor (MC1R), which is stably overexpressed in the majority of primary malignant melanomas, and integrin αvβ3, which is involved in lymph node metastasis and therefore has an important role in the transition from local to metastatic disease, are important target receptors. Thus, if a radiolabeled HBPL could be developed that was able to bind to both receptor types, the early diagnosis and correct staging of the disease would be significantly increased. Here, we report on the design, synthesis, radiolabeling and in vitro and in vivo testing of different SiFAlin-modified HBPLs (SiFA = silicon fluoride acceptor), consisting of an MC1R-targeting (GG-Nle-c(DHfRWK)) and an integrin αvβ3-affine peptide (c(RGDfK)), being connected by a symmetrically branching framework including linkers of differing length and composition. Kit-like 18F-radiolabeling of the HBPLs 1–6 provided the labeled products [18F]1–[18F]6 in radiochemical yields of 27–50%, radiochemical purities of ≥95% and non-optimized molar activities of 17–51 GBq/μmol within short preparation times of 25 min. Besides the evaluation of radiotracers regarding logD(7.4) and stability in human serum, the receptor affinities of the HBPLs were investigated in vitro on cell lines overexpressing integrin αvβ3 (U87MG cells) or the MC1R (B16F10). Based on these results, the most promising compounds [18F]2, showing the highest affinity to both target receptors (IC50 (B16F10) = 0.99 ± 0.11 nM, IC50 (U87MG) = 1300 ± 288 nM), and [18F]4, exhibiting the highest hydrophilicity (logD(7.4) = −1.39 ± 0.03), were further investigated in vivo and ex vivo in a xenograft mouse model bearing both tumors. For both HBPLs, clear visualization of B16F10, as well as U87MG tumors, was feasible. Blocking studies using the respective monospecific peptides demonstrated both peptide binders of the HBPLs contributing to tumor uptake. Despite the somewhat lower target receptor affinities (IC50 (B16F10) = 6.00 ± 0.47 nM and IC50 (U87MG) = 2034 ± 323 nM) of [18F]4, the tracer showed higher absolute tumor uptakes ([18F]4: 2.58 ± 0.86% ID/g in B16F10 tumors and 3.92 ± 1.31% ID/g in U87MG tumors; [18F]2: 2.32 ± 0.49% ID/g in B16F10 tumors and 2.33 ± 0.46% ID/g in U87MG tumors) as well as higher tumor-to-background ratios than [18F]2. Thus, [18F]4 demonstrates to be a highly potent radiotracer for the sensitive and bispecific imaging of malignant melanoma by PET/CT imaging and impressively illustrates the suitability of the underlying concept to develop heterobivalent integrin αvβ3- and MC1R-bispecific radioligands for the sensitive and specific imaging of malignant melanoma by PET/CT.
Collapse
Affiliation(s)
- Xia Cheng
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany; (X.C.); (V.v.K.)
| | - Ralph Hübner
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany;
| | - Valeska von Kiedrowski
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany; (X.C.); (V.v.K.)
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany;
| | - Ralf Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada;
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany;
- Correspondence: (C.W.); (B.W.)
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany; (X.C.); (V.v.K.)
- Correspondence: (C.W.); (B.W.)
| |
Collapse
|
6
|
Current State of Radiolabeled Heterobivalent Peptidic Ligands in Tumor Imaging and Therapy. Pharmaceuticals (Basel) 2020; 13:ph13080173. [PMID: 32751666 PMCID: PMC7465997 DOI: 10.3390/ph13080173] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
Over the past few years, an approach emerged that combines different receptor-specific peptide radioligands able to bind different target structures on tumor cells concomitantly or separately. The reason for the growing interest in this special field of radiopharmaceutical development is rooted in the fact that bispecific peptide heterodimers can exhibit a strongly increased target cell avidity and specificity compared to their corresponding monospecific counterparts by being able to bind to two different target structures that are overexpressed on the cell surface of several malignancies. This increase of avidity is most pronounced in the case of concomitant binding of both peptides to their respective targets but is also observed in cases of heterogeneously expressed receptors within a tumor entity. Furthermore, the application of a radiolabeled heterobivalent agent can solve the ubiquitous problem of limited tumor visualization sensitivity caused by differential receptor expression on different tumor lesions. In this article, the concept of heterobivalent targeting and the general advantages of using radiolabeled bispecific peptidic ligands for tumor imaging or therapy as well as the influence of molecular design and the receptors on the tumor cell surface are explained, and an overview is given of the radiolabeled heterobivalent peptides described thus far.
Collapse
|
7
|
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: 2.3] [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
|
8
|
Gaonkar RH, Baishya R, Paul B, Dewanjee S, Ganguly S, Debnath MC, Ganguly S. Development of a peptide-based bifunctional chelator conjugated to a cytotoxic drug for the treatment of melanotic melanoma. MEDCHEMCOMM 2018; 9:812-826. [PMID: 30108970 PMCID: PMC6071751 DOI: 10.1039/c7md00638a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/28/2018] [Indexed: 01/02/2023]
Abstract
The cytotoxic drug gemcitabine (GEM) has been conjugated to receptor-binding peptides to target melanoma tumors. A hexapeptide having a Lys-Gly-His-Lys sequence (pep-1), an octapeptide with an Arg-Gly-Asp-Lys-Gly-His-Lys sequence (pep-2), a GEM-conjugated Lys-Gly-His-Lys peptide (GEM-pep-3) and a GEM-conjugated Asp-Gly-Arg peptide (GEM-pep-4) were synthesized and characterized. In vitro uptake of fluorescently labeled GEM-pep-3 and GEM-pep-4 on B16F10 cells was investigated. Fluorescence microscopy studies demonstrated significant uptake of GEM-pep-3 in the B16F10 mouse melanoma cell line. The peptides and GEM-coupled peptides were radiolabeled with [99mTc(CO)3(H2O)3]+ and examined for in vitro cell binding in the B16F10 melanoma cell line and in vivo biodistribution and scintigraphic studies in a B16F10 melanoma tumor-bearing mice model. In vitro cellular uptake studies and biological evaluation confirmed significant deposition of GEM-pep-3 at the melanoma tumor site. The MTT assay depicted higher cytotoxic behaviour of GEM-pep-3 than free GEM. A considerable amount of cell apoptosis was also observed in B16F10 cells. Finally, the in vivo therapeutic efficacy study revealed a significant decrease in tumor growth in the GEM-pep-3-treated animal model. These studies reveal enough potentiality of GEM-pep-3 to treat melanoma and underline the need for further evaluation.
Collapse
Affiliation(s)
- Raghuvir H Gaonkar
- Infectious Diseases and Immunology Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India .
| | - Rinku Baishya
- Natural Product Chemistry Group , Chemical Science and Technology Division , North East Institute of Science and Technology , Assam , India
| | - Brahamacharry Paul
- Infectious Diseases and Immunology Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India .
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory , Department of Pharmaceutical Technology , Jadavpur University , Kolkata , India
| | - Shantanu Ganguly
- Regional Radiation Medicine Center , Thakurpukur Cancer Center and Welfare Home Campus , Kolkata , India
| | - Mita C Debnath
- Infectious Diseases and Immunology Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India .
| | - Soumya Ganguly
- Infectious Diseases and Immunology Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India .
| |
Collapse
|
9
|
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: 3.7] [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
|
10
|
Ericson MD, Lensing CJ, Fleming KA, Schlasner KN, Doering SR, Haskell-Luevano C. Bench-top to clinical therapies: A review of melanocortin ligands from 1954 to 2016. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2414-2435. [PMID: 28363699 PMCID: PMC5600687 DOI: 10.1016/j.bbadis.2017.03.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
The discovery of the endogenous melanocortin agonists in the 1950s have resulted in sixty years of melanocortin ligand research. Early efforts involved truncations or select modifications of the naturally occurring agonists leading to the development of many potent and selective ligands. With the identification and cloning of the five known melanocortin receptors, many ligands were improved upon through bench-top in vitro assays. Optimization of select properties resulted in ligands adopted as clinical candidates. A summary of every melanocortin ligand is outside the scope of this review. Instead, this review will focus on the following topics: classic melanocortin ligands, selective ligands, small molecule (non-peptide) ligands, ligands with sex-specific effects, bivalent and multivalent ligands, and ligands advanced to clinical trials. Each topic area will be summarized with current references to update the melanocortin field on recent progress. This article is part of a Special Issue entitled: Melanocortin Receptors - edited by Ya-Xiong Tao.
Collapse
Affiliation(s)
- Mark D Ericson
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Cody J Lensing
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katlyn A Fleming
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katherine N Schlasner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Skye R Doering
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
11
|
de Oliveira EA, Lazovic J, Guo L, Soto H, Faintuch BL, Akhtari M, Pope W. Evaluation of Magnetonanoparticles Conjugated with New Angiogenesis Peptides in Intracranial Glioma Tumors by MRI. Appl Biochem Biotechnol 2017; 183:265-279. [PMID: 28281182 DOI: 10.1007/s12010-017-2443-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/20/2017] [Indexed: 01/12/2023]
Abstract
Angiogenesis plays a critical role in progression of malignant gliomas. The development of glioma-specific labeling molecules that can aid detection and visualization of angiogenesis can help surgical planning and improve treatment outcome. The aim of this study was to evaluate if two peptides (GX1 and RGD-GX1) linked to angiogenesis can be used as an MR-imaging markers of angiogenesis. MR imaging was performed in U87 glioblastoma-bearing NOD-SCID mice at different time points between 15 and 120 min post-injection to visualize particle distribution. GX1 and RGD-GX1 exhibited the highest accumulation in U87 glioblastoma at 120 min post i.v. administration. GX1-conjugated agents lead to higher decrease in transverse relaxation time (T 2) (i.e., stronger contrast enhancement) than RGD-GX1-conjugated agents in U87 glioblastoma tumor model. In addition, we tested if U87-IDH1R132 mutated cell line had different pattern of GX1 or RGD-GX1 particle accumulation. Responses in U87-IDH1WT followed a similar pattern with GX1 contrast agents; however, lower contrast enhancement was observed with RGD-GX1 agents. The specific binding of these peptides to human glioblastoma xenograft in the brain was confirmed by magnetic resonance imaging. The contrast enhancement following injection of magnetonanoparticles conjugated to GX1 peptide matched well with CD31 staining and iron staining.
Collapse
Affiliation(s)
- Erica Aparecida de Oliveira
- Radiopharmacy Center, Institute of Energy and Nuclear Research, Av. Prof. Lineu Prestes 2242, São Paulo, SP, 05508-000, Brazil. .,School of Pharmaceutical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 580 Bloco 17, São Paulo, SP, 05508-900, Brazil.
| | - Jelena Lazovic
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lea Guo
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Horacio Soto
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Bluma Linkowski Faintuch
- Radiopharmacy Center, Institute of Energy and Nuclear Research, Av. Prof. Lineu Prestes 2242, São Paulo, SP, 05508-000, Brazil
| | - Massoud Akhtari
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen school of Medicine, University of California, Los Angeles, CA, USA
| | - Whitney Pope
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
12
|
Tc-99m-labeled RGD-conjugated alpha-melanocyte stimulating hormone hybrid peptides with reduced renal uptake. Amino Acids 2015; 47:813-23. [PMID: 25557051 DOI: 10.1007/s00726-014-1911-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/24/2014] [Indexed: 10/24/2022]
Abstract
The purpose of this study was to examine whether the replacement of the positively-charged Lys or Arg linker with a neutral linker could reduce the renal uptake of Arg-Gly-Asp (RGD)-conjugated alpha-melanocyte stimulating hormone (α-MSH) hybrid peptide. The RGD motif {cyclic(Arg-Gly-Asp-DTyr-Asp)} was coupled to [Cys(3,4,10), D-Phe(7), Arg(11)]α-MSH3-13 {(Arg(11))CCMSH} through the neutral βAla or Ahx {aminohexanoic acid} linker (replacing the Lys or Arg linker) to generate novel RGD-βAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH hybrid peptides. The receptor-binding affinity and cytotoxicity of RGD-βAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH were determined in B16/F1 melanoma cells. The melanoma targeting and imaging properties of (99m)Tc-RGD-βAla-(Arg(11))CCMSH and (99m)Tc-RGD-Ahx-(Arg(11))CCMSH were determined in B16/F1 melanoma-bearing C57 mice. The replacement of the Lys or Arg linker with the βAla or Ahx linker retained nanomolar receptor-binding affinities and remarkable cytotoxicity of RGD-βAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH. The receptor-binding affinities of RGD-βAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH were 0.8 ± 0.05 and 1.3 ± 0.1 nM. Three-hour incubation with 0.1 µM of RGD-βAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH decreased the survival percentages of B16/F1 cells by 71 and 67 % as compared to the untreated control cells 5 days post the treatment. The replacement of the Arg linker with the βAla or Ahx linker reduced the non-specific renal uptake of (99m)Tc-RGD-βAla-(Arg(11))CCMSH and (99m)Tc-RGD-Ahx-(Arg(11))CCMSH by 62 and 61 % at 2 h post-injection. (99m)Tc-RGD-βAla-(Arg(11))CCMSH displayed higher melanoma uptake than (99m)Tc-RGD-Ahx-(Arg(11))CCMSH at 0.5, 2, 4, and 24 h post-injection. Enhanced tumor to kidney uptake ratio of (99m)Tc-RGD-βAla-(Arg(11))CCMSH warranted the further evaluation of (188)Re-labeled RGD-βAla-(Arg(11))CCMSH as a novel MC1 receptor-targeting therapeutic peptide for melanoma treatment in the future.
Collapse
|
13
|
Parat A, Kryza D, Degoul F, Taleb J, Viallard C, Janier M, Garofalo A, Bonazza P, Heinrich-Balard L, Cohen R, Miot-Noirault E, Chezal JM, Billotey C, Felder-Flesch D. Radiolabeled dendritic probes as tools for high in vivo tumor targeting: application to melanoma. J Mater Chem B 2015; 3:2560-2571. [DOI: 10.1039/c5tb00235d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A small-sized and bifunctional111In-radiolabeled dendron shows highin vivotargeting efficiency towards an intracellular target in a murine melanoma model.
Collapse
|
14
|
Oliveira EA, Faintuch BL. Radiolabeling and biological evaluation of the GX1 and RGD-GX1 peptide sequence for angiogenesis targeting. Nucl Med Biol 2014; 42:123-30. [PMID: 25311749 DOI: 10.1016/j.nucmedbio.2014.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/03/2014] [Accepted: 09/15/2014] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Aiming to develop a novel (99m)Tc-labeled imaging agent, for angiogenesis and tumor receptors, two peptides obtained from phage display library, namely GX1 and the heterodimer RGD-GX1, were synthesized in a cyclic conformation. They were radiolabeled with (99m)Tc, employing the HYNIC chelator, for radiochemical evaluation and biological properties. METHODS Radiolabeling, radiochemical control, plasma protein binding, and partition coefficient were assessed for both radioconjugates. Biodistribution in healthy Balb/c mice was carried out, in order to evaluate the biological behaviour of the radiocomplexes. RESULTS The conjugates displayed a rather similar pharmacokinetic profile. They were prepared with high radiochemical purity (>96%), and both were hydrophilic (log P of -2.25 and -2.51 respectively). Preferential renal excretion was observed. Kidney uptake (42.31±5.35 %ID/g) for (99m)Tc-HYNIC-E-[c(RGDfk)-c(GX1)], 1h post-injection was about three times higher than the uptake of (99m)Tc-HYNIC-PEG4-c(GX1) (11.92±4.77%ID/g). Total blood, bone and muscle values revealed a slightly slower clearance for the RGD-GX1 radiocomplex. CONCLUSION The high radiochemical purity achieved, and the similar in vivo profile observed for both radioconjugates, make them potential candidates for radiopharmaceuticals for tumor imaging. Further investigations of binding affinity, and uptake of GX1 and RGD-GX1 peptides in tumor models, are warranted.
Collapse
Affiliation(s)
- E A Oliveira
- Radiopharmacy, Institute of Energy and Nuclear Research, Sao Paulo, SP, Brazil, Av. Prof. Lineu Prestes, 2242 05508-000 São Paulo, SP, Brazil.
| | - B L Faintuch
- Radiopharmacy, Institute of Energy and Nuclear Research, Sao Paulo, SP, Brazil, Av. Prof. Lineu Prestes, 2242 05508-000 São Paulo, SP, Brazil
| |
Collapse
|
15
|
Yang J, Flook AM, Feng C, Miao Y. Linker modification reduced the renal uptake of technetium-99m-labeled Arg-Ala-Asp-conjugated alpha-melanocyte stimulating hormone peptide. Bioorg Med Chem Lett 2014; 24:195-8. [PMID: 24316121 PMCID: PMC3889140 DOI: 10.1016/j.bmcl.2013.11.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 11/13/2013] [Accepted: 11/15/2013] [Indexed: 11/30/2022]
Abstract
The purpose of this study was to examine the biodistribution of (99m)Tc-RAD-Arg-(Arg(11))CCMSH in B16/F1 melanoma-bearing C57 mice to determine whether the replacement of the Lys linker with an Arg linker could decrease the renal uptake of (99m)Tc-RAD-Arg-(Arg(11))CCMSH. (99m)Tc-RAD-Arg-(Arg(11))CCMSH exhibited rapid and high tumor uptake (17.98±4.96% ID/g at 2h post-injection) in B16/F1 melanoma-bearing C57 mice. As compared to (99m)Tc-RAD-Lys-(Arg(11))CCMSH, the replacement of the Lys linker with an Arg linker dramatically decreased the renal uptake of (99m)Tc-RAD-Arg-(Arg(11))CCMSH by 68%, 62%, 73% and 64% at 0.5, 2, 4 and 24h post-injection, respectively. Flank B16/F1 melanoma lesions were clearly imaged at 2h post-injection using (99m)Tc-RAD-Arg-(Arg(11))CCMSH as an imaging probe.
Collapse
Affiliation(s)
- Jianquan Yang
- 2502 Marble NE, MSC09 5360, College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Adam M Flook
- 2502 Marble NE, MSC09 5360, College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Changjian Feng
- 2502 Marble NE, MSC09 5360, College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Yubin Miao
- 2502 Marble NE, MSC09 5360, College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA; Cancer Research and Treatment Center, University of New Mexico, Albuquerque, NM 87131, USA; Department of Dermatology, University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
16
|
Flook AM, Yang J, Miao Y. Effects of amino acids on melanoma targeting and clearance properties of Tc-99m-labeled Arg-X-Asp-conjugated α-melanocyte stimulating hormone peptides. J Med Chem 2013; 56:8793-802. [PMID: 24131154 DOI: 10.1021/jm4012356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The purpose of this study was to examine the effects of amino acids on melanoma targeting and clearance properties of new (99m)Tc-labeled Arg-X-Asp-conjugated α-melanocyte stimulating hormone (α-MSH) peptides. RSD-Lys-(Arg(11))CCMSH {c[Arg-Ser-Asp-DTyr-Asp]-Lys-Cys-Cys-Glu-His-dPhe-Arg-Trp-Cys-Arg-Pro-Val-NH2}, RNleD-Lys-(Arg(11))CCMSH, RPheD-Lys-(Arg(11))CCMSH, and RdPheD-Lys-(Arg(11))CCMSH peptides were synthesized and evaluated for their melanocortin-1 (MC1) receptor binding affinities in B16/F1 melanoma cells. The biodistribution of (99m)Tc-RSD-Lys-(Arg(11))CCMSH, (99m)Tc-RFD-Lys-(Arg(11))CCMSH, and (99m)Tc-RfD-Lys-(Arg(11))CCMSH were determined in B16/F1 melanoma-bearing C57 mice. The substitution of Gly with Ser, Phe, and dPhe increased the MC1 receptor binding affinities of the peptides, whereas the substitution of Gly with Nle decreased the MC1 receptor binding affinity of the peptide. (99m)Tc-RSD-Lys-(Arg(11))CCMSH exhibited the highest melanoma uptake (18.01 ± 4.22% ID/g) and the lowest kidney and liver uptake among these (99m)Tc-peptides. The B16/F1 melanoma lesions could be clearly visualized by SPECT/CT using (99m)Tc-RSD-Lys-(Arg(11))CCMSH as an imaging probe. It is desirable to reduce the renal uptake of (99m)Tc-RSD-Lys-(Arg(11))CCMSH to facilitate its potential therapeutic application.
Collapse
Affiliation(s)
- Adam M Flook
- College of Pharmacy, ‡Cancer Research and Treatment Center, §Department of Dermatology, University of New Mexico , Albuquerque, New Mexico, 87131, United States
| | | | | |
Collapse
|
17
|
Rosenkranz AA, Slastnikova TA, Durymanov MO, Sobolev AS. Malignant melanoma and melanocortin 1 receptor. BIOCHEMISTRY. BIOKHIMIIA 2013; 78:1228-37. [PMID: 24460937 PMCID: PMC4064721 DOI: 10.1134/s0006297913110035] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The conventional chemotherapeutic treatment of malignant melanoma still remains poorly efficient in most cases. Thus the use of specific features of these tumors for development of new therapeutic modalities is highly needed. Melanocortin 1 receptor (MC1R) overexpression on the cell surface of the vast majority of human melanomas, making MC1R a valuable marker of these tumors, is one of these features. Naturally, MC1R plays a key role in skin protection against damaging ultraviolet radiation by regulating eumelanin production. MC1R activation is involved in regulation of melanocyte cell division. This article reviews the peculiarities of regulation and expression of MC1R, melanocytes, and melanoma cells, along with the possible connection of MC1R with signaling pathways regulating proliferation of tumor cells. MC1R is a cell surface endocytic receptor, thus considered perspective for diagnostics and targeted drug delivery. A number of new therapeutic approaches that utilize MC1R, including endoradiotherapy with Auger electron and α- and β-particle emitters, photodynamic therapy, and gene therapy are now being developed.
Collapse
Affiliation(s)
- A. A. Rosenkranz
- Institute of Gene Biology, Russian Academy of Sciences, ul. Vavilova 34/5, 199334 Moscow, Russia; fax: +7 (499) 135-4105
- Faculty of Biology, Lomonosov Moscow State University, Leninsky Gory 1-12, 119234 Moscow, Russia; fax: +7 (495) 939-4309;
- Targeted Delivery of Pharmaceuticals “Translek” LLC, ul. Vavilova 34/5, 199334 Moscow, Russia;
| | - T. A. Slastnikova
- Institute of Gene Biology, Russian Academy of Sciences, ul. Vavilova 34/5, 199334 Moscow, Russia; fax: +7 (499) 135-4105
| | - M. O. Durymanov
- Institute of Gene Biology, Russian Academy of Sciences, ul. Vavilova 34/5, 199334 Moscow, Russia; fax: +7 (499) 135-4105
- Faculty of Biology, Lomonosov Moscow State University, Leninsky Gory 1-12, 119234 Moscow, Russia; fax: +7 (495) 939-4309;
| | - A. S. Sobolev
- Institute of Gene Biology, Russian Academy of Sciences, ul. Vavilova 34/5, 199334 Moscow, Russia; fax: +7 (499) 135-4105
- Faculty of Biology, Lomonosov Moscow State University, Leninsky Gory 1-12, 119234 Moscow, Russia; fax: +7 (495) 939-4309;
- Targeted Delivery of Pharmaceuticals “Translek” LLC, ul. Vavilova 34/5, 199334 Moscow, Russia;
| |
Collapse
|
18
|
Flook AM, Yang J, Miao Y. Evaluation of new Tc-99m-labeled Arg-X-Asp-conjugated α-melanocyte stimulating hormone peptides for melanoma imaging. Mol Pharm 2013; 10:3417-24. [PMID: 23885640 DOI: 10.1021/mp400248f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The purpose of this study was to examine the melanoma targeting and imaging properties of two new (99m)Tc-labeled Arg-X-Asp-conjugated α-melanocyte stimulating hormone (α-MSH) peptides. RTD-Lys-(Arg(11))CCMSH {c[Asp-Arg-Thr-Asp-DTyr]-Lys-Cys-Cys-Glu-His-DPhe-Arg-Trp-Cys-Arg-Pro-Val-NH2} and RVD-Lys-(Arg(11))CCMSH peptides were synthesized, and their melanocortin-1 (MC1) receptor binding affinities were determined in B16/F1 melanoma cells. The biodistribution and melanoma imaging properties of (99m)Tc-RTD-Lys-(Arg(11))CCMSH and (99m)Tc-RVD-Lys-(Arg(11))CCMSH were determined in B16/F1 melanoma-bearing C57 mice. The IC50 values of RTD-Lys-(Arg(11))CCMSH and RVD-Lys-(Arg(11))CCMSH were 0.7 ± 0.07 and 1.0 ± 0.3 nM in B16/F1 melanoma cells. Both (99m)Tc-RTD-Lys-(Arg(11))CCMSH and (99m)Tc-RVD-Lys-(Arg(11))CCMSH displayed high melanoma uptake. (99m)Tc-RTD-Lys-(Arg(11))CCMSH exhibited the highest tumor uptake of 18.77 ± 5.13% ID/g at 2 h postinjection, whereas (99m)Tc-RVD-Lys-(Arg(11))CCMSH reached the highest tumor uptake of 19.63 ± 4.68% ID/g at 4 h postinjection. Both (99m)Tc-RTD-Lys-(Arg(11))CCMSH and (99m)Tc-RVD-Lys-(Arg(11))CCMSH showed low accumulation in normal organs (<1.7% ID/g) except for the kidneys at 2 h postinjection. The renal uptake of (99m)Tc-RTD-Lys-(Arg(11))CCMSH and (99m)Tc-RVD-Lys-(Arg(11))CCMSH was 135.14 ± 23.62 and 94.01 ± 18.31% ID/g at 2 h postinjection, respectively. The melanoma lesions were clearly visualized by single-photon emission computed tomography (SPECT)/CT using either (99m)Tc-RTD-Lys-(Arg(11))CCMSH or (99m)Tc-RVD-Lys-(Arg(11))CCMSH as an imaging probe at 2 h postinjection. Overall, the introduction of Thr or Val residue retained high melanoma uptake of (99m)Tc-RTD-Lys-(Arg(11))CCMSH and (99m)Tc-RVD-Lys-(Arg(11))CCMSH. However, high renal uptake of (99m)Tc-RTD-Lys-(Arg(11))CCMSH and (99m)Tc-RVD-Lys-(Arg(11))CCMSH need to be reduced to facilitate their future applications.
Collapse
Affiliation(s)
- Adam M Flook
- College of Pharmacy, ‡Cancer Research and Treatment Center, and §Department of Dermatology, University of New Mexico , Albuquerque, New Mexico 87131, United States
| | | | | |
Collapse
|
19
|
Fischer G, Schirrmacher R, Wängler B, Wängler C. Radiolabeled Heterobivalent Peptidic Ligands: an Approach with High Future Potential for in vivo Imaging and Therapy of Malignant Diseases. ChemMedChem 2013; 8:883-90. [DOI: 10.1002/cmdc.201300081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Indexed: 12/19/2022]
|
20
|
Gene therapy for advanced melanoma: selective targeting and therapeutic nucleic acids. JOURNAL OF DRUG DELIVERY 2013; 2013:897348. [PMID: 23634303 PMCID: PMC3619548 DOI: 10.1155/2013/897348] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 02/24/2013] [Indexed: 12/21/2022]
Abstract
Despite recent advances, the treatment of malignant melanoma still results in the relapse of the disease, and second line treatment mostly fails due to the occurrence of resistance. A wide range of mutations are known to prevent effective treatment with chemotherapeutic drugs. Hence, approaches with biopharmaceuticals including proteins, like antibodies or cytokines, are applied. As an alternative, regimens with therapeutically active nucleic acids offer the possibility for highly selective cancer treatment whilst avoiding unwanted and toxic side effects. This paper gives a brief introduction into the mechanism of this devastating disease, discusses the shortcoming of current therapy approaches, and pinpoints anchor points which could be harnessed for therapeutic intervention with nucleic acids. We bring the delivery of nucleic acid nanopharmaceutics into perspective as a novel antimelanoma therapeutic approach and discuss the possibilities for melanoma specific targeting. The latest reports on preclinical and already clinical application of nucleic acids in melanoma are discussed.
Collapse
|
21
|
Koopmans KP, Glaudemans AWJM. Rationale for the use of radiolabelled peptides in diagnosis and therapy. Eur J Nucl Med Mol Imaging 2012; 39 Suppl 1:S4-10. [PMID: 22388630 DOI: 10.1007/s00259-011-2038-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Nuclear medicine techniques are becoming more important in imaging oncological and infectious diseases. For metabolic imaging of these diseases, antibody and peptide imaging are currently used. In recent years peptide imaging has become important, therefore the rationale for the use of peptide imaging is described in this article. Criteria for a successful peptide tracer are a high target specificity, a high binding affinity, a long metabolic stability and a high target-to-background ratio. Tracer internalization is also beneficial. For oncological imaging, many tracers are available, most originating from regulatory peptides, but penetrating peptides are also being developed. Peptides for imaging inflammatory and infectious diseases include regulatory peptides, antimicrobial peptides and others. In conclusion, for the imaging of oncological, imflammatory and infectious diseases, many promising peptides are being developed. The ideal peptide probe is characterized by rapid and specific target localization and binding with a high tumour-to-background ratio.
Collapse
Affiliation(s)
- K P Koopmans
- Department of Radiology and Nuclear Medicine, Martini Hospital, Van Swietenplein 1, 9728 NT Groningen, The Netherlands.
| | | |
Collapse
|
22
|
Yang J, Lu J, Miao Y. Structural modification on the Lys linker enhanced tumor to kidney uptake ratios of 99mTc-labeled RGD-conjugated α-MSH hybrid peptides. Mol Pharm 2012; 9:1418-24. [PMID: 22452443 DOI: 10.1021/mp2006642] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The purpose of this study was to examine whether the structural modification on the positively charged Lys linker could reduce the kidney uptake of (99m)Tc-labeled Arg-Gly-Asp (RGD)-conjugated α-melanocyte stimulating hormone (α-MSH) hybrid peptides. The RGD motif {cyclic(Arg-Gly-Asp-D-Tyr-Asp)} was coupled to [Cys(3,4,10), D-Phe(7), Arg(11)]α-MSH(3-13) {(Arg(11))CCMSH} through a neutral glycine linker to eliminate the positively charged amino side chain of the Lys linker or without a linker to delete the Lys linker. The receptor binding affinity of RGD-Gly-(Arg(11))CCMSH and RGD-(Arg(11))CCMSH was determined in B16/F1 melanoma cells. The melanoma targeting and imaging properties of (99m)Tc-RGD-Gly-(Arg(11))CCMSH and (99m)Tc-RGD-(Arg(11))CCMSH were determined in B16/F1 melanoma-bearing C57 mice. The structural modification on the Lys linker retained a low nanomolar receptor binding affinity of RGD-Gly-(Arg(11))CCMSH and RGD-(Arg(11))CCMSH (1.5 and 1.0 nM, respectively). The structural modification on the Lys linker dramatically decreased the renal uptake of (99m)Tc-RGD-Gly-(Arg(11))CCMSH and (99m)Tc-RGD-(Arg(11))CCMSH by 79% and 77% at 4 h postinjection compared to (99m)Tc-RGD-Lys-(Arg(11))CCMSH. (99m)Tc-RGD-(Arg(11))CCMSH displayed a higher melanoma uptake (16.12 ± 3.09% ID/g) than (99m)Tc-RGD-Gly-(Arg(11))CCMSH (11.50 ± 1.01% ID/g) at 2 postinjection. The tumor uptake of (99m)Tc-RGD-(Arg(11))CCMSH was 1.4 times the tumor uptake of (99m)Tc-RGD-Gly-(Arg(11))CCMSH at 2 postinjection. A dramatically enhanced tumor-to-kidney uptake ratio of (99m)Tc-RGD-(Arg(11))CCMSH suggests that (188)Re-RGD-(Arg(11))CCMSH may behave in a similar fashion warranting future evaluation for melanoma treatment.
Collapse
Affiliation(s)
- Jianquan Yang
- College of Pharmacy, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | | | | |
Collapse
|
23
|
Yang J, Miao Y. Substitution of Gly with Ala enhanced the melanoma uptake of technetium-99m-labeled Arg-Ala-Asp-conjugated alpha-melanocyte stimulating hormone peptide. Bioorg Med Chem Lett 2012; 22:1541-5. [PMID: 22297112 DOI: 10.1016/j.bmcl.2012.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 01/03/2012] [Accepted: 01/03/2012] [Indexed: 11/28/2022]
Abstract
The purpose of this study was to determine the melanoma targeting property of (99m)Tc-RAD-Lys-(Arg(11))CCMSH in B16/F1 melanoma-bearing C57 mice and compare with (99m)Tc-RGD-Lys-(Arg(11))CCMSH we previously reported. (99m)Tc-RAD-Lys-(Arg(11))CCMSH exhibited rapid and high tumor uptake (19.91±4.02% ID/g at 2h post-injection) in B16/F1 melanoma-bearing C57 mice. The tumor uptake of (99m)Tc-RAD-Lys-(Arg(11))CCMSH was 1.51, 1.34 and 1.43 times the tumor uptake of (99m)Tc-RGD-Lys-(Arg(11))CCMSH at 0.5, 2 and 4h post-injection, respectively. Flank B16/F1 melanoma lesions were clearly imaged at 2h post-injection using (99m)Tc-RAD-Lys-(Arg(11))CCMSH as an imaging probe. The substitution of Gly with Ala significantly enhanced the melanoma uptake of (99m)Tc-RAD-Lys-(Arg(11))CCMSH compared to (99m)Tc-RGD-Lys-(Arg(11))CCMSH in B16/F1 melanoma-bearing C57 mice, providing a new insight into the design of α-MSH peptides for melanoma targeting.
Collapse
Affiliation(s)
- Jianquan Yang
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | | |
Collapse
|
24
|
Baumhover NJ, Martin ME, Parameswarappa SG, Kloepping KC, O'Dorisio MS, Pigge FC, Schultz MK. Improved synthesis and biological evaluation of chelator-modified α-MSH analogs prepared by copper-free click chemistry. Bioorg Med Chem Lett 2011; 21:5757-61. [PMID: 21873053 DOI: 10.1016/j.bmcl.2011.08.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 07/30/2011] [Accepted: 08/02/2011] [Indexed: 01/23/2023]
Abstract
Radionuclide chelators (DOTA, NOTA) functionalized with a monofluorocyclooctyne group were prepared. These materials reacted rapidly and in high yield with a fully deprotected azide-modified peptide via Cu-free click chemistry under mild reaction conditions (aqueous solution, room temperature). The resulting bioconjugates bind with high affinity and specificity to their cell-surface receptor targets in vitro and appear stable to degradation in mouse serum over 3h of incubation at 37°C.
Collapse
Affiliation(s)
- Nicholas J Baumhover
- Department of Radiology, Carver College of Medicine, The University of Iowa, 500 Newton Road, ML B180, Iowa City, IA 52242, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Gomes CM, Abrunhosa AJ, Ramos P, Pauwels EKJ. Molecular imaging with SPECT as a tool for drug development. Adv Drug Deliv Rev 2011; 63:547-54. [PMID: 20933557 DOI: 10.1016/j.addr.2010.09.015] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 09/22/2010] [Accepted: 09/28/2010] [Indexed: 01/10/2023]
Abstract
Molecular imaging techniques are increasingly being used as valuable tools in the drug development process. Radionuclide-based imaging modalities such as single-photon emission computed tomography (SPECT) and positron emission tomography (PET) have proven to be useful in phases ranging from preclinical development to the initial stages of clinical testing. The high sensitivity of these imaging modalities makes them particularly suited for exploratory investigational new drug (IND) studies as they have the potential to characterize in vivo pharmacokinetics and biodistribution of the compounds using only a fraction of the intended therapeutic dose (microdosing). This information obtained at an early stage of clinical testing results in a better selection among promising drug candidates, thereby increasing the success rate of agents entering clinical trials and the overall efficiency of the process. In this article, we will review the potential applications of SPECT imaging in the drug development process with an emphasis on its applications in exploratory IND studies.
Collapse
Affiliation(s)
- Célia M Gomes
- Institute of Biophysics/Biomathematics - IBILI, Faculty of Medicine, Coimbra University, Portugal.
| | | | | | | |
Collapse
|
26
|
|
27
|
Correia JDG, Paulo A, Raposinho PD, Santos I. Radiometallated peptides for molecular imaging and targeted therapy. Dalton Trans 2011; 40:6144-67. [DOI: 10.1039/c0dt01599g] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
28
|
Current World Literature. Curr Opin Support Palliat Care 2010; 4:293-304. [DOI: 10.1097/spc.0b013e328340e983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Yan Y, Chen K, Yang M, Sun X, Liu S, Chen X. A new 18F-labeled BBN-RGD peptide heterodimer with a symmetric linker for prostate cancer imaging. Amino Acids 2010; 41:439-47. [PMID: 20936525 DOI: 10.1007/s00726-010-0762-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 09/23/2010] [Indexed: 12/13/2022]
Abstract
A peptide heterodimer comprises two different receptor-targeting peptide ligands. Molecular imaging probes based on dual-receptor targeting peptide heterodimers exhibit improved tumor targeting efficacy for multi-receptor expressing tumors compared with their parent single-receptor targeting peptide monomers. Previously we have developed bombesin (BBN)-RGD (Arg-Gly-Asp) peptide heterodimers, in which BBN and RGD are covalently connected with an asymmetric glutamate linker (J Med Chem 52:425-432, 2009). Although (18)F-labeled heterodimers showed significantly better microPET imaging quality than (18)F-labeled RGD and BBN monomers in a PC-3 xenograft model which co-expresses gastrin-releasing peptide receptor (GRPR) and integrin αvβ3, tedious heterodimer synthesis due to the asymmetric nature of glutamate linker restricts their clinical applications. In this study, we report the use of a symmetric linker AEADP [AEADP = 3,3'-(2-aminoethylazanediyl)dipropanoic acid] for the synthesis of BBN-RGD peptide heterodimer. The (18)F-labeled heterodimer ((18)F-FB-AEADP-BBN-RGD) showed comparable microPET imaging results with glutamate linked BBN-RGD heterodimers, indicating that the replacement of glutamate linker with AEADP linker did not affect the biological activities of BBN-RGD heterodimer. The heterodimer synthesis is rather easy and straightforward. Because tumors often co-express multiple receptors, the use of a symmetric linker provides a general method of fast assembly of various peptide heterodimers for imaging multi-receptor expressing tumors.
Collapse
Affiliation(s)
- Yongjun Yan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 31 Center Drive, Suite 1C14, Bethesda, MD 20892-2281, USA
| | | | | | | | | | | |
Collapse
|
30
|
Lee S, Xie J, Chen X. Peptides and peptide hormones for molecular imaging and disease diagnosis. Chem Rev 2010; 110:3087-111. [PMID: 20225899 DOI: 10.1021/cr900361p] [Citation(s) in RCA: 256] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Seulki Lee
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 31 Center Drive, Suite 1C14, Bethesda, Maryland 20892-2281, USA
| | | | | |
Collapse
|
31
|
Replacement of the Lys linker with an Arg linker resulting in improved melanoma uptake and reduced renal uptake of Tc-99m-labeled Arg-Gly-Asp-conjugated alpha-melanocyte stimulating hormone hybrid peptide. Bioorg Med Chem 2010; 18:6695-700. [PMID: 20728365 DOI: 10.1016/j.bmc.2010.07.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 07/20/2010] [Accepted: 07/27/2010] [Indexed: 11/23/2022]
Abstract
The purpose of this study was to reduce the non-specific renal uptake of Arg-Gly-Asp (RGD)-conjugated alpha-melanocyte stimulating hormone (alpha-MSH) hybrid peptide through structural modification or L-lysine co-injection. The RGD motif {cyclic(Arg-Gly-Asp-DTyr-Asp)} was coupled to [Cys(3,4,10), D-Phe7, Arg11] alpha-MSH3-13 {(Arg11)CCMSH} through the Arg linker (substituting the Lys linker) to generate a novel RGD-Arg-(Arg11)CCMSH hybrid peptide. The melanoma targeting and pharmacokinetic properties of 99mTc-RGD-Arg-(Arg11)CCMSH were determined in B16/F1 melanoma-bearing C57 mice. The effect of L-lysine co-injection on the renal uptake was determined through the co-injection of L-lysine with 99mTc-RGD-Arg-(Arg11)CCMSH or 99mTc-RGD-Lys-(Arg11)CCMSH. Replacement of the Lys linker with an Arg linker exhibited a profound effect in reducing the non-specific renal uptake of 99mTc-RGD-Arg-(Arg11)CCMSH, as well as increasing the tumor uptake of 99mTc-RGD-Arg-(Arg11)CCMSH compared to 99mTc-RGD-Lys-(Arg11)CCMSH. 99mTc-RGD-Arg-(Arg11)CCMSH exhibited high tumor uptake (21.41+/-3.74% ID/g at 2 h post-injection) and prolonged tumor retention (6.81+/-3.71% ID/g at 24 h post-injection) in B16/F1 melanoma-bearing mice. The renal uptake values of 99mTc-RGD-Arg-(Arg11)CCMSH were 40.14-64.08% of those of 99mTc-RGD-Lys-(Arg11)CCMSH (p<0.05) at 0.5, 2, 4 and 24 h post-injection. Co-injection of L-lysine was effective in decreasing the renal uptakes of 99mTc-RGD-Arg-(Arg11)CCMSH by 27.7% and 99mTc-RGD-Lys-(Arg11)CCMSH by 52.1% at 2 h post-injection. Substitution of the Lys linker with an Arg linker dramatically improved the melanoma uptake and reduced the renal uptake of 99mTc-RGD-Arg-(Arg11)CCMSH, warranting the further evaluation of 188Re-labeled RGD-Arg-(Arg11)CCMSH as a novel MC1 receptor-targeting therapeutic peptide for melanoma treatment in the future.
Collapse
|
32
|
Yang J, Guo H, Miao Y. Technetium-99m-labeled Arg-Gly-Asp-conjugated alpha-melanocyte stimulating hormone hybrid peptides for human melanoma imaging. Nucl Med Biol 2010; 37:873-83. [PMID: 21055617 DOI: 10.1016/j.nucmedbio.2010.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 05/07/2010] [Accepted: 05/17/2010] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The purpose of this study was to examine whether (99m)Tc-labeled Arg-Gly-Asp (RGD)-conjugated alpha-melanocyte stimulating hormone (α-MSH) hybrid peptide targeting both melanocortin-1 (MC1) and α(v)β(3) integrin receptors was superior in melanoma targeting to (99m)Tc-labeled α-MSH or RGD peptide targeting only the MC1 or α(v)β(3) integrin receptor. METHODS RGD-Lys-(Arg(11))CCMSH, RAD-Lys-(Arg(11))CCMSH and RGD-Lys-(Arg(11))CCMSHscramble were designed to target both MC1 and α(v)β(3) integrin receptors, MC1 receptor only and α(v)β(3) integrin receptor only, respectively. The MC1 or α(v)β(3) integrin receptor binding affinities of three peptides were determined in M21 human melanoma cells. The melanoma targeting properties of (99m)Tc-labeled RGD-Lys-(Arg(11))CCMSH, RAD-Lys-(Arg(11))CCMSH and RGD-Lys-(Arg(11))CCMSHscramble were determined in M21 human melanoma-xenografted nude mice. Meanwhile, the melanoma uptake of (99m)Tc-RGD-Lys-(Arg(11))CCMSH was blocked with various non-radiolabeled peptides in M21 melanoma xenografts. RESULTS RGD-Lys-(Arg(11))CCMSH displayed 2.0 and 403 nM binding affinities to both MC1 and α(v)β(3) integrin receptors, whereas RAD-Lys-(Arg(11))CCMSH or RGD-Lys-(Arg(11))CCMSHscramble lost their α(v)β(3) integrin receptor binding affinity by greater than 248-fold or MC1 receptor binding affinity by more than 100-fold, respectively. The melanoma uptake of (99m)Tc-RGD-Lys-(Arg(11))CCMSH was 2.49 and 2.24 times (P < .05) the melanoma uptakes of (99m)Tc-RAD-Lys-(Arg(11))CCMSH and (99m)Tc-RGD-Lys-(Arg(11))CCMSHscramble at 2 h post-injection, respectively. Either RGD or (Arg(11))CCMSH peptide co-injection could block 42% and 57% of the tumor uptake of (99m)Tc-RGD-Lys-(Arg(11))CCMSH, whereas the coinjection of RGD+(Arg(11))CCMSH peptide mixture could block 66% of the tumor uptake of (99m)Tc-RGD-Lys-(Arg(11))CCMSH. CONCLUSIONS Targeting both MC1 and α(v)β(3) integrin receptors enhanced the melanoma uptake of (99m)Tc-RGD-Lys-(Arg(11))CCMSH in M21 human melanoma xenografts. Flank M21 human melanoma tumors were clearly visualized by single photon emission computed tomography/computed tomographic imaging using (99m)Tc-RGD-Lys-(Arg(11))CCMSH as an imaging probe, highlighting its potential use as a dual-receptor-targeting imaging probe for human melanoma detection.
Collapse
Affiliation(s)
- Jianquan Yang
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
33
|
Quinn T, Zhang X, Miao Y. Targeted melanoma imaging and therapy with radiolabeled alpha-melanocyte stimulating hormone peptide analogues. GIORN ITAL DERMAT V 2010; 145:245-58. [PMID: 20467398 PMCID: PMC2999912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Radiolabeled alpha-melanocyte stimulating hormone (a-MSH) analogues have been used to define the expression, affinity and function of the melanocortin-1 receptor (MC1-R). The MC1-R is one of a family of five G-protein linker receptors, which is primarily involved in regulation of skin pigmentation. Over-expression of the MC1-R on melanoma tumor cells has made it an attractive target for the development of a-MSH peptide based imaging and therapeutic agents. Initially, the native a-MSH peptide was radiolabeled directly, but it suffered from low specific activity and poor stability. The addition of non-natural amino acids yielded a-MSH analogues with greater MC-1R affinity and stability. Furthermore, peptide cyclization via disulfide and lactam bond formation as well as site-specific metal coordination resulted in additional gains in receptor affinity and peptide stability in vitro and in vivo. Radiochemical stability of the a-MSH analogues was improved through the conjugation of metal chelators to the peptide's N-terminus or lysine residues for radionuclide coordination. In vitro cell binding studies demonstrated that the radiolabeled a-MSH analogues had low to subnanomolar affinities for the MC1-R. Biodistribution and imaging studies in the B16 mouse melanoma modeled showed rapid tumor uptake of the radiolabeled peptides, with the cyclic peptides demonstrating prolonged tumor retention. Cyclic a-MSH analogues labeled with beta and alpha emitting radionuclides demonstrated melanoma therapeutic efficacy in the B16 melanoma mouse model. Strong pre-clinical imaging and therapy data highlight the clinical potential use of radiolabeled a-MSH peptides for melanoma imaging and treatment of disseminated disease.
Collapse
Affiliation(s)
- T Quinn
- Biochemistry Department, University of Missouri, Harry S Truman Memorial Veterans Administration Hospital Columbia, MO 65211, USA.
| | | | | |
Collapse
|
34
|
Nanda PK, Lane SR, Retzloff LB, Pandey US, Smith CJ. Radiolabeled regulatory peptides for imaging and therapy. Curr Opin Endocrinol Diabetes Obes 2010; 17:69-76. [PMID: 19901831 PMCID: PMC2892114 DOI: 10.1097/med.0b013e32833392ac] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW The purpose of the present review is to describe new, innovative strategies of diagnosing and treating specific human cancers using a cadre of radiolabeled regulatory peptides. RECENT FINDINGS Peptide receptor-targeted radionuclide therapy is a method of site-directed radiotherapy that specifically targets human cancers expressing a cognate receptor-subtype in very high numbers. Ideally, the procedure targets only the primary or metastatic disease and is minimally invasive, with little radiation damage to normal, collateral tissues. For treatment strategies of this type to be effective, it is critical to evaluate the toxicity of the treatment protocol, the radiation dosimetry of the therapeutic regimen, and the biological profile of the radiopharmaceutical, including biodistribution and pharmacokinetics of the drug. Site-directed molecular imaging procedures via gamma-scintigraphy can address many of the critical issues associated with peptide receptor-targeted radionuclide therapy and it is, therefore, necessary to describe the effective balance between the clinical benefits and risks of this treatment strategy. SUMMARY Continued development in the design or chemical structure of radiolabeled, biologically active peptides could do much to improve the targeting ability of these drugs, thereby creating new and innovative strategies for diagnosis or treatment of human cancers.
Collapse
Affiliation(s)
- Prasant K. Nanda
- Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Stephanie R. Lane
- Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Department of Chemistry, University of Missouri, Columbia, Missouri, USA
| | - Lauren B. Retzloff
- Department of Molecular Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Usha S. Pandey
- Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Charles Jeffrey Smith
- Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Research Division, Harry S. Truman Memorial Veterans’ Hospital, University of Missouri School of Medicine, Columbia, Missouri, USA
- Department of Molecular Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- The Radiopharmaceutical Sciences Institute, University of Missouri School of Medicine, Columbia, Missouri, USA
- University of Missouri Research Reactor Center, Columbia, Missouri, USA
| |
Collapse
|