1
|
Erfani M, Mikaeili A, Fallah Z, Goudarzi M. Preclinical evaluation of a new technetium-99m labeled neurotensin analogue for NTSR1 targeted radionuclide imaging. Bioorg Chem 2024; 153:107858. [PMID: 39395320 DOI: 10.1016/j.bioorg.2024.107858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/15/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
Neurotensin is a regulatory peptide that can act as a growth factor on different types of normal and cancerous cells. Binding of Neurotensin to relevant receptors leads to cell proliferation, survival, migration and invasion by changing intracellular enzyme activity. Therefore, the design of a neurotensin-based radiopeptide plays an important role in targeted imaging or therapy of neurotensin receptor-positive tumors. A [Lys8]-neurotensin (7-13) peptide was synthesized and attached to HYNIC as a chelator via a linker. The labeling procedure was carried out at 100 °C for 10 min using 99mTc as a radionuclide and EDDA/tricine as coligands. Stability of the labeled peptide in human serum was determined using RTLC and HPLC methods. The receptor binding internalization was studied using HT-29 colon carcinoma cells, and tissue biodistribution was evaluated in mice bearing CT-26 tumors. The [99mTc]Tc-Tricine/EDDA/HYNIC-GABA-[Lys8]-neurotensin (7-13) peptide demonstrated a labeling yield of over 98 %, a specific activity of 37.00 GBq/µmol, high stability in human serum, a nanomolar range of Kd, and a tumor uptake of 0.36 ± 0.15 % ID/g at 1-h post-injection. These results suggest that the labeled peptide is a suitable imaging agent for neurotensin receptor-positive tumors.
Collapse
Affiliation(s)
- Mostafa Erfani
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran.
| | - Azadeh Mikaeili
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | - Zhila Fallah
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | - Mostafa Goudarzi
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| |
Collapse
|
2
|
Previti S, Bodin S, Rémond E, Vimont D, Hindié E, Morgat C, Cavelier F. Rational design of NT-PSMA heterobivalent probes for prostate cancer theranostics. RSC Med Chem 2024:d4md00491d. [PMID: 39371434 PMCID: PMC11451938 DOI: 10.1039/d4md00491d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/12/2024] [Indexed: 10/08/2024] Open
Abstract
Targeting the prostate-specific membrane antigen (PSMA) with radiopharmaceuticals for imaging and/or therapy has demonstrated significant advancement in the management of prostate cancer patients. However, PSMA targeting remains unsuccessful in prostate cancers with low expression of PSMA, which account for 15% of cases. The neurotensin receptor-1 (NTS1) has been highlighted as a suitable oncotarget for imaging and therapy of PSMA-negative prostate cancer lesions. Therefore, heterobivalent probes targeting both PSMA and NTS1 could improve the prostate cancer management. Herein, we report the development of a branched hybrid probe (JMV 7489) designed to target PSMA and/or NTS1 bearing relevant pharmacophores and DOTA as the chelating agent. The new ligand was synthesized with a hybrid approach, which includes both syntheses in batch and in the solid phase. Saturation binding experiments were next performed on HT-29 and PC3-PIP cells to derive K d and B max values. On the PC3-PIP cells, [68Ga]Ga-JMV 7489 displayed good affinity towards PSMA (K d = 53 ± 17 nM; B max = 1393 ± 29 fmol/106 cells) in the same range as the corresponding reference monomer. A lower affinity value towards NTS1 was depicted (K d = 157 ± 71 nM; B max = 241 ± 42 fmol/106 cells on PC3-PIP cells; K d = 246 ± 1 nM; B max = 151 ± 44 fmol/106 cells on HT-29 cells) and, surprisingly, it was also the case for the corresponding monomer [68Ga]Ga-JMV 7089. These results indicate that the DOTA macrocycle and the linker are critical elements to design heterobivalent probes targeting PSMA and NTS1 with high affinity towards NTS1.
Collapse
Affiliation(s)
- Santo Previti
- Pôle Chime Balard, IBMM, UMR 5247 CNRS, Université Montpellier ENSCM F-34000 Montpellier France +33 448792134
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina Viale Stagno d'Alcontres 31 98166 Messina Italy +39 090 676 5669
| | - Sacha Bodin
- CNRS, EPHE, INCIA UMR 5287, University of Bordeaux F-33400 Talence France
- Department of Nuclear Medicine, CHU Bordeaux F-33000 Bordeaux France
| | - Emmanuelle Rémond
- Pôle Chime Balard, IBMM, UMR 5247 CNRS, Université Montpellier ENSCM F-34000 Montpellier France +33 448792134
| | - Delphine Vimont
- CNRS, EPHE, INCIA UMR 5287, University of Bordeaux F-33400 Talence France
| | - Elif Hindié
- CNRS, EPHE, INCIA UMR 5287, University of Bordeaux F-33400 Talence France
- Department of Nuclear Medicine, CHU Bordeaux F-33000 Bordeaux France
- Institut Universitaire de France F-75000 Paris France
| | - Clément Morgat
- CNRS, EPHE, INCIA UMR 5287, University of Bordeaux F-33400 Talence France
- Department of Nuclear Medicine, CHU Bordeaux F-33000 Bordeaux France
| | - Florine Cavelier
- Pôle Chime Balard, IBMM, UMR 5247 CNRS, Université Montpellier ENSCM F-34000 Montpellier France +33 448792134
| |
Collapse
|
3
|
Fan W, Zhang W, Allen S, Alshehri S, Muilenburg KM, Zheng C, Garrison JC. Examination of Charge Modifications of an Endolysosomal Trapping Inhibitor in an Antagonistic NTSR1-Targeted Construct for Colon Cancer. Bioconjug Chem 2022; 33:1363-1376. [PMID: 35793523 PMCID: PMC9941984 DOI: 10.1021/acs.bioconjchem.2c00214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Many low-molecular weight targeted radiotherapeutics (TRTs) are capable of rapidly achieving exceptional tumor to non-target ratios shortly after administration. However, the low tumor residence time of many TRTs limits therapeutic dose delivery and has become the Achilles heel to their clinical translation. To combat the tumor efflux of these otherwise promising agents, we have previously presented a strategy of equipping low-molecular weight TRTs with irreversible cysteine cathepsin inhibitors (e.g., E-64 analogues). These inhibitors are capable of forming irreversible adducts with cysteine proteases within the endolysosomal compartments of cells. Using these endolysosomal trapping agents (ETs), the receptor-targeted constructs are able to increase tumor retention and, thus, deliverable therapeutic doses. In this study, we examine this approach in the development of agents targeting the neurotensin receptor subtype 1 (NTSR1), a receptor overexpressed in numerous cancers. Using an antagonistic NTSR1-targeting vector, we explore the impact of charge modification of the ETs on the in vitro and in vivo biological performance of the constructs using HT-29 colon cancer models. Four ETs (based on the epoxysuccinyl peptide E-64) with various charge states were synthesized and incorporated into the structures of the NTSR1-targeted antagonist. These four 177Lu-labeled, ET-enhanced, NTSR1-targeted agents (177Lu-NA-ET1-4), along with the structurally analogous 177Lu-3BP-227, currently in clinical trials, underwent a battery of in vitro assays using HT-29 xenograft colon cancer cells to examine their NTSR1 binding, internalization and efflux, inhibition, and adduct formation properties. The biodistribution profile of these constructs was studied in an HT-29 mouse model. Charge modification of the terminal carboxylic acid and arginine of the ETs had deleterious effects on inhibition kinetics and in vitro adduct formation. Contrastingly, deletion of the arginine resulted in a modest increase in inhibition kinetics. Incorporation of ETs into the NTSR1-targeted agents was well-tolerated with minimal impact on the in vivo NTSR1 targeting but resulted in increased renal uptake. This study demonstrates that the ETs can be successfully incorporated into antagonistic NTSR1-targeted constructs without compromising their adduct formation capabilities. Based on these results, further exploration of the endolysosomal trapping approach is warranted in NTSR1- and other receptor-targeted antagonistic constructs.
Collapse
Affiliation(s)
- Wei Fan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE
| | - Wenting Zhang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE
| | - Sadie Allen
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE
| | - Sameer Alshehri
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE
| | - Kathryn M Muilenburg
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE
| | - Cheng Zheng
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE
| | - Jered C. Garrison
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE,Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE,Corresponding Author Jered C. Garrison, Tel: +01 4025593453.
| |
Collapse
|
4
|
Fan W, Zhang W, Alshehri S, Garrison JC. Examination of the impact molecular charge has on NTSR1-targeted agents incorporated with cysteine protease inhibitors. Eur J Med Chem 2022; 234:114241. [PMID: 35306289 PMCID: PMC9007894 DOI: 10.1016/j.ejmech.2022.114241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 02/05/2023]
Abstract
Our laboratory has previously reported a strategy of employing cysteine cathepsin (CC) inhibitors as adduct forming, trapping agents to extend the tumor residence time of neurotensin receptor subtype 1 (NTSR1)-targeted radiopharmaceuticals. As a follow-up, we herein report a small library of CC trapping agent (CCTA)-incorporated, NTSR1-targeted conjugates with structural modifications that reduce the number of charged functional groups for both the CCTA and the peptide targeting sequence. These modifications were pursued to reduce the renal uptake and increase the translational potential of the CCTA-incorporated, NTSR1-targeted agents as radiotherapeutics. The biological performance of these constructs was examined using a battery of in vitro and in vivo studies employing the NTSR1-positive HT-29 human colon cancer cell line as our model. In vitro studies confirmed the ability of these constructs to target the NTSR1 and efficiently form intracellular adducts with cysteine proteases. Biodistribution studies using an HT-29 xenograft mouse model revealed that truncation (removal of Lys6-Pro7) of the NTSR1-targeted peptide (177Lu-NE2a) had the greatest (3.7-fold) effect at lowering renal recognition/uptake relative to our previously reported construct. Other charge-reducing modifications to the CCTA resulted in unexpected increases in renal uptake. All of the constructs demonstrated similar levels of in vivo NTSR1-positive tumor targeting with the highest tumor residualization resulting from the construct containing the zwitterionic CCTA (177Lu-NE2a). In vivo adduct formation of the conjugates was confirmed using autoradiographic SDS-PAGE analysis.
Collapse
Affiliation(s)
- Wei Fan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, United States,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, United States
| | - Wenting Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, United States,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, United States
| | - Sameer Alshehri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, United States,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, United States
| | - Jered C. Garrison
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, United States,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, United States,Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE 68198, United States,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, NE 68105, United States,Corresponding author: Tel: +01 4025593453,
| |
Collapse
|
5
|
Effendi N, Mishiro K, Shiba K, Kinuya S, Ogawa K. Development of Radiogallium-Labeled Peptides for Platelet-Derived Growth Factor Receptor β (PDGFR β) Imaging: Influence of Different Linkers. Molecules 2020; 26:molecules26010041. [PMID: 33374773 PMCID: PMC7795354 DOI: 10.3390/molecules26010041] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
The purpose of this study is to develop peptide-based platelet-derived growth factor receptor β (PDGFRβ) imaging probes and examine the effects of several linkers, namely un-natural amino acids (D-alanine and β-alanine) and ethylene-glycol (EG), on the properties of Ga-DOTA-(linker)-IPLPPPRRPFFK peptides. Seven radiotracers, 67Ga-DOTA-(linker)-IPLPPPRRPFFK peptides, were designed, synthesized, and evaluated. The stability and cell uptake in PDGFRβ positive peptide cells were evaluated in vitro. The biodistribution of [67Ga]Ga-DOTA-EG2-IPLPPPRRPFFK ([67Ga]27) and [67Ga]Ga-DOTA-EG4-IPLPPPRRPFFK ([67Ga]28), which were selected based on in vitro stability in murine plasma and cell uptake rates, were determined in BxPC3-luc-bearing nu/nu mice. Seven 67Ga-labeled peptides were successfully synthesized with high radiochemical yields (>85%) and purities (>99%). All evaluated radiotracers were stable in PBS (pH 7.4) at 37 °C. However, only [67Ga]27 and [67Ga]28 remained more than 75% after incubation in murine plasma at 37 °C for 1 h. [67Ga]27 exhibited the highest BxPC3-luc cell uptake among the prepared radiolabeled peptides. As regards the results of the biodistribution experiments, the tumor-to-blood ratios of [67Ga]27 and [67Ga]28 at 1 h post-injection were 2.61 ± 0.75 and 2.05 ± 0.77, respectively. Co-injection of [67Ga]27 and an excess amount of IPLPPPRRPFFK peptide as a blocking agent can significantly decrease this ratio. However, tumor accumulation was not considered sufficient. Therefore, further probe modification is required to assess tumor accumulation for in vivo imaging.
Collapse
Affiliation(s)
- Nurmaya Effendi
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan; (N.E.); (K.M.)
- Faculty of Pharmacy, Universitas Muslim Indonesia, Urip Sumiharjo KM. 10, Makassar 90-231, Indonesia
| | - Kenji Mishiro
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan; (N.E.); (K.M.)
| | - Kazuhiro Shiba
- Advanced Science Research Center, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa 920-8640, Japan;
| | - Seigo Kinuya
- Department of Nuclear Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa 920-8641, Japan;
| | - Kazuma Ogawa
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan; (N.E.); (K.M.)
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
- Correspondence: ; Tel./Fax: +81-76-234-4460
| |
Collapse
|
6
|
Kanellopoulos P, Nock BA, Krenning EP, Maina T. Optimizing the Profile of [ 99mTc]Tc-NT(7-13) Tracers in Pancreatic Cancer Models by Means of Protease Inhibitors. Int J Mol Sci 2020; 21:ijms21217926. [PMID: 33114537 PMCID: PMC7663772 DOI: 10.3390/ijms21217926] [Citation(s) in RCA: 3] [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: 09/26/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 12/17/2022] Open
Abstract
Background: The overexpression of neurotensin subtype 1 receptors (NTS1Rs) in human tumors may be elegantly exploited for directing neurotensin (NT)-based radionuclide carriers specifically to cancer sites for theranostic purposes. We have recently shown that [99mTc]Tc–DT1 ([99mTc]Tc–[N4–Gly7]NT(7–13)) and [99mTc]Tc–DT5 ([99mTc]Tc–[N4–βAla7,Dab9]NT(7–13)) show notably improved uptake in human colon adenocarcinoma WiDr xenografts in mice treated with neprilysin (NEP) inhibitors and/or angiotensin-converting enzyme (ACE) inhibitors compared with untreated controls. Aiming toward translation of this promising approach in NTS1R-positive pancreatic ductal adenocarcinoma (PDAC) patients, we now report on the impact of registered NEP/ACE inhibitors on the performance of [99mTc]Tc–DT1 and [99mTc]Tc–DT5 in pancreatic cancer models. Methods: The cellular uptake of [99mTc]Tc–DT1 and [99mTc]Tc–DT5 was tested in a panel of pancreatic cell lines, and their stability was assessed in mice treated or not treated with Entresto, lisinopril, or their combinations. Biodistribution was conducted in severe combined immunodeficiency (SCID) mice bearing pancreatic AsPC-1 xenografts. Results: The Entresto + lisinopril combination maximized the metabolic stability of the fast-internalizing [99mTc]Tc–DT1 in mice, resulting in notably enhanced tumor uptake (7.05 ± 0.80% injected activity (IA)/g vs. 1.25 ± 0.80% IA/g in non-treated controls at 4 h post-injection; p < 0.0001). Conclusions: This study has shown the feasibility of optimizing the uptake of [99mTc]Tc–DT1 in pancreatic cancer models with the aid of clinically established NEP/ACE inhibitors, in favor of clinical translation prospects.
Collapse
Affiliation(s)
- Panagiotis Kanellopoulos
- Molecular Radiopharmacy, INRASTES, NCSR “Demokritos”, 15341 Athens, Greece;
- Molecular Pharmacology, School of Medicine, University of Crete, Heraklion, 70013 Crete, Greece
- Correspondence: (P.K.); (T.M.); Tel.: +30-210-650-3891 (P.K.); +30-210-650-3908 (T.M.)
| | - Berthold A. Nock
- Molecular Radiopharmacy, INRASTES, NCSR “Demokritos”, 15341 Athens, Greece;
| | - Eric P. Krenning
- Cyclotron Rotterdam BV, Erasmus MC, 3015 CE Rotterdam, The Netherlands;
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR “Demokritos”, 15341 Athens, Greece;
- Correspondence: (P.K.); (T.M.); Tel.: +30-210-650-3891 (P.K.); +30-210-650-3908 (T.M.)
| |
Collapse
|
7
|
Li D, Minnix M, Allen R, Bading J, Chea J, Wong P, Bowles N, Poku E, Shively JE. Preclinical PET Imaging of NTSR-1-Positive Tumors with 64Cu- and 68Ga-DOTA-Neurotensin Analogs and Therapy with an 225Ac-DOTA-Neurotensin Analog. Cancer Biother Radiopharm 2020; 36:651-661. [PMID: 32822229 DOI: 10.1089/cbr.2020.3926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Background: The aim of the study was to perform PET imaging and radiotherapy with a novel neurotensin derivative for neurotensin receptor 1 (NTSR-1)-positive tumors in an animal model. Materials and Methods: A di-DOTA analog of NT(6-13) with three unnatural amino acids was synthesized and radiolabeled with either 64Cu or 68Ga and tested for serum stability and tumor imaging in mice bearing NTSR-1-positive PC3, and HT29 xenografts. A dose-response therapy study was performed with 18.5, 37, and 74 kBq of 225Ac-di-DOTA-α,ɛ-Lys-NT(6-13). Results: 68Ga-di-DOTA-α,ɛ-Lys-NT(6-13) was >99% stable in serum for 48 h, had an IC50 of 5 nM using 125I labeled NT(8-13) for binding to HT-29 cells, and high uptake in tumor models expressing NTSR-1. 68Ga-di-DOTA-α,ɛ-Lys-NT(6-13) had an average %ID/g (n = 4) at 2 h of 4.0 for tumor, 0.5 for blood, 12.0 for kidney, and <1 for other tissues, resulting in a favorable T/B of 8. Mean survivals of tumor-bearing mice treated with 18.5 or 37 kBq of 225Ac-di-DOTA-α,ɛ-Lys-NT(6-13) were 81 and 93 d, respectively, versus 53 d for controls. Whole-body toxicity was seen for the 74 kBq dose. Conclusions: Based on the results of the animal model, di-DOTA-α,ɛ-Lys-NT(6-13) is a useful imaging agent for NTSR-1-positive tumors when radiolabeled with 68Ga, and when radiolabeled with 225Ac, a potent therapeutic agent.
Collapse
Affiliation(s)
- Daneng Li
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - Megan Minnix
- Deparment of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Rebecca Allen
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - James Bading
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - Junie Chea
- Radiopharmacy, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | - Patty Wong
- Deparment of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Nicole Bowles
- Radiopharmacy, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | - Erasmus Poku
- Radiopharmacy, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | - John E Shively
- Deparment of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| |
Collapse
|
8
|
Kanellopoulos P, Kaloudi A, de Jong M, Krenning EP, Nock BA, Maina T. Key-Protease Inhibition Regimens Promote Tumor Targeting of Neurotensin Radioligands. Pharmaceutics 2020; 12:pharmaceutics12060528. [PMID: 32526874 PMCID: PMC7356968 DOI: 10.3390/pharmaceutics12060528] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023] Open
Abstract
Neurotensin subtype 1 receptors (NTS1R) represent attractive molecular targets for directing radiolabeled neurotensin (NT) analogs to tumor lesions for diagnostic and therapeutic purposes. This approach has been largely undermined by the rapid in vivo degradation of linear NT-based radioligands. Herein, we aim to increase the tumor targeting of three 99mTc-labeled NT analogs by the in-situ inhibition of two key proteases involved in their catabolism. DT1 ([N4-Gly7]NT(7-13)), DT5 ([N4-βAla7,Dab9]NT(7-13)), and DT6 ([N4-βAla7,Dab9,Tle12]]NT(7-13)) were labeled with 99mTc. Their profiles were investigated in NTS1R-positive colon adenocarcinoma WiDr cells and mice treated or not with the neprilysin (NEP)-inhibitor phosphoramidon (PA) and/or the angiotensin converting enzyme (ACE)-inhibitor lisinopril (Lis). Structural modifications led to the partial stabilization of 99mTc-DT6 in peripheral mice blood (55.1 ± 3.9% intact), whereas 99mTc-DT1 and 99mTc-DT5 were totally degraded within 5 min. Coinjection of PA and/or Lis significantly stabilized all three analogs, leading to a remarkable enhancement of tumor uptake for 99mTc-DT1 and 99mTc-DT5, but was less effective in the case of poorly internalizing 99mTc-DT6. In conclusion, NEP and/or ACE inhibition represents a powerful tool to improve tumor targeting and the overall pharmacokinetics of NT-based radioligands, and warrants further validation in the field of NTS1R-targeted tumor imaging and therapy.
Collapse
Affiliation(s)
- Panagiotis Kanellopoulos
- Molecular Radiopharmacy, INRASTES, NCSR “Demokritos”, 15341 Athens, Greece; (P.K.); (A.K.); (B.A.N.)
- Molecular Pharmacology, School of Medicine, University of Crete, 70013 Heraklion, Greece
| | - Aikaterini Kaloudi
- Molecular Radiopharmacy, INRASTES, NCSR “Demokritos”, 15341 Athens, Greece; (P.K.); (A.K.); (B.A.N.)
| | - Marion de Jong
- Department of Radiology & Nuclear Medicine Erasmus MC, 3015 CN Rotterdam, The Netherlands;
| | - Eric P. Krenning
- Cyclotron Rotterdam BV, Erasmus MC, 3015 CE Rotterdam, The Netherlands;
| | - Berthold A. Nock
- Molecular Radiopharmacy, INRASTES, NCSR “Demokritos”, 15341 Athens, Greece; (P.K.); (A.K.); (B.A.N.)
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR “Demokritos”, 15341 Athens, Greece; (P.K.); (A.K.); (B.A.N.)
- Correspondence: ; Tel.: +30-210-650-3908
| |
Collapse
|
9
|
Structural modifications of amino acid sequences of radiolabeled peptides for targeted tumor imaging. Bioorg Chem 2020; 99:103802. [DOI: 10.1016/j.bioorg.2020.103802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 12/18/2022]
|
10
|
Farahani AM, Maleki F, Sadeghzadeh N. The Influence of Different Spacers on Biological Profile of Peptide Radiopharmaceuticals for Diagnosis and Therapy of Human Cancers. Anticancer Agents Med Chem 2020; 20:402-416. [PMID: 31889492 DOI: 10.2174/1871520620666191231161227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/07/2019] [Accepted: 12/12/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Cancer is the leading cause of death worldwide. Early detection can reduce the disadvantageous effects of diseases and the mortality in cancer. Nuclear medicine is a powerful tool that has the ability to diagnose malignancy without harming normal tissues. In recent years, radiolabeled peptides have been investigated as potent agents for cancer detection. Therefore, it is necessary to modify radiopeptides in order to achieve more effective agents. OBJECTIVE This review describes modifications in the structure of radioconjugates with spacers who have improved the specificity and sensitivity of the peptides that are used in oncologic diagnosis and therapy. METHODS To improve the biological activity, researchers have conjugated these peptide analogs to different spacers and bifunctional chelators. Many spacers of different kinds, such as hydrocarbon chain, amino acid sequence, and poly (ethyleneglycol) were introduced in order to modify the pharmacokinetic properties of these biomolecules. RESULTS Different spacers have been applied to develop radiolabeled peptides as potential tracers in nuclear medicine. Spacers with different charge and hydrophilicity affect the characteristics of peptide conjugate. For example, the complex with uncharged and hydrophobic spacers leads to increased liver uptake, while the composition with positively charged spacers results in high kidney retention. Therefore, the pharmacokinetics of radio complexes correlates to the structure and total charge of the conjugates. CONCLUSION Radio imaging technology has been successfully applied to detect a tumor in the earliest stage. For this purpose, the assessment of useful agents to diagnose the lesion is necessary. Developing peptide radiopharmaceuticals using spacers can improve in vitro and in vivo behavior of radiotracers leading to better noninvasive detection and monitoring of tumor growth.
Collapse
Affiliation(s)
- Arezou M Farahani
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Mazandaran, Sari, Iran.,Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fariba Maleki
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Mazandaran, Sari, Iran.,Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nourollah Sadeghzadeh
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Mazandaran, Sari, Iran
| |
Collapse
|
11
|
Schindler L, Bernhardt G, Keller M. Modifications at Arg and Ile Give Neurotensin(8-13) Derivatives with High Stability and Retained NTS 1 Receptor Affinity. ACS Med Chem Lett 2019; 10:960-965. [PMID: 31223455 DOI: 10.1021/acsmedchemlett.9b00122] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Due to its expression in various malignant tumors, the neurotensin receptor 1 (NTS1R) has been suggested and explored as a target for tumor diagnosis and therapy. Animal model-based investigations of various radiolabeled NTS1R ligands derived from the hexapeptide neurotensin(8-13) (NT(8-13)), e.g. 68Ga- and 18F-labeled compounds for PET diagnostics, give rise to optimize such radiotracers for clinical use. As NT(8-13) is rapidly degraded in vivo; structural modifications are required in terms of increased metabolic stability. In this study, the stabilization of the peptide backbone of NT(8-13) against enzymatic degradation was systematically explored by performing an N-methyl scan, replacing Ile12 by tert-butylglycine12 (Tle12) and N-terminal acylation. N-Methylation of either arginine, Arg8, or Arg9, combined with the Ile12/Tle12 exchange, proved to be most favorable with respect to NTS1R affinity (K i < 2 nM) and stability in human plasma (t 1/2 > 48 h), a valuable result regarding the development of radiopharmaceuticals derived from NT(8-13).
Collapse
Affiliation(s)
- Lisa Schindler
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| |
Collapse
|
12
|
Fan W, Zhang W, Alshehri S, Neeley TR, Garrison JC. Enhanced tumor retention of NTSR1-targeted agents by employing a hydrophilic cysteine cathepsin inhibitor. Eur J Med Chem 2019; 177:386-400. [PMID: 31158752 DOI: 10.1016/j.ejmech.2019.05.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 01/09/2023]
Abstract
We explored the approach of using an analog of E-64, a well-known and hydrophilic cysteine cathepsin (CC) inhibitor, as a potent cysteine cathepsin-trapping agent (CCTA) to improve the tumor retention of low-molecular-weight, receptor-targeted radiopharmaceuticals. The synthesized hydrophilic CCTA-incorporated, NTSR1-targeted agents demonstrated a substantial increase in cellular retention upon uptake into the NTRS1-positive HT-29 human colon cancer cell line. Similarly, biodistribution studies using HT-29 xenograft mice revealed a significant and substantial increase in tumor retention for the CCTA-incorporated, NTSR1-targeted agent. The intracellular trapping mechanism of the CCTA-incorporated agents by macromolecular adduct formation was confirmed using multiple in vitro and in vivo techniques. Furthermore, utilization of the more hydrophilic CCTA greatly increased the hydrophilicity of the resulting NTSR1-targeted constructs leading to substantial decreases in most non-target tissues in contrast to our previously reported dipeptidyl acyloxymethyl ketone (AOMK) constructs. This work further confirms that the CCTA trapping approach can make significant improvements in the clinical potential of NTSR1-and other receptor-targeted radiopharmaceuticals.
Collapse
Affiliation(s)
- Wei Fan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States; Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Wenting Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States; Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Sameer Alshehri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States; Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Trey R Neeley
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States; Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Jered C Garrison
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States; Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, 68198, United States; Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, United States; Eppley Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, NE, 68198, United States.
| |
Collapse
|
13
|
Fan W, Zhang W, Alshehri S, Garrison JC. Increasing time on target: utilization of inhibitors of cysteine cathepsins to enhance the tumor retention of receptor-targeted agents. Chem Commun (Camb) 2018; 54:11268-11271. [PMID: 30182093 DOI: 10.1039/c8cc05982a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We report a strategy of utilizing irreversible cysteine cathepsin inhibitor as trapping agent to increase the tumor residence time of receptor-targeted agents. The targeted constructs incorporating these cysteine cathepsin trapping agents were able to form high molecular weight adducts with intracellular cysteine cathepsins, thus achieving superior retention in tumor tissues.
Collapse
Affiliation(s)
- Wei Fan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198, USA.
| | | | | | | |
Collapse
|
14
|
Debordeaux F, Chansel-Debordeaux L, Pinaquy JB, Fernandez P, Schulz J. What about αvβ3 integrins in molecular imaging in oncology? Nucl Med Biol 2018; 62-63:31-46. [DOI: 10.1016/j.nucmedbio.2018.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/19/2018] [Accepted: 04/30/2018] [Indexed: 10/17/2022]
|
15
|
Korbecki J, Gutowska I, Kojder I, Jeżewski D, Goschorska M, Łukomska A, Lubkowska A, Chlubek D, Baranowska-Bosiacka I. New extracellular factors in glioblastoma multiforme development: neurotensin, growth differentiation factor-15, sphingosine-1-phosphate and cytomegalovirus infection. Oncotarget 2018; 9:7219-7270. [PMID: 29467963 PMCID: PMC5805549 DOI: 10.18632/oncotarget.24102] [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] [Received: 07/12/2017] [Accepted: 01/02/2018] [Indexed: 11/25/2022] Open
Abstract
Recent years have seen considerable progress in understanding the biochemistry of cancer. For example, more significance is now assigned to the tumor microenvironment, especially with regard to intercellular signaling in the tumor niche which depends on many factors secreted by tumor cells. In addition, great progress has been made in understanding the influence of factors such as neurotensin, growth differentiation factor-15 (GDF-15), sphingosine-1-phosphate (S1P), and infection with cytomegalovirus (CMV) on the 'hallmarks of cancer' in glioblastoma multiforme. Therefore, in the present work we describe the influence of these factors on the proliferation and apoptosis of neoplastic cells, cancer stem cells, angiogenesis, migration and invasion, and cancer immune evasion in a glioblastoma multiforme tumor. In particular, we discuss the effect of neurotensin, GDF-15, S1P (including the drug FTY720), and infection with CMV on tumor-associated macrophages (TAM), microglial cells, neutrophil and regulatory T cells (Treg), on the tumor microenvironment. In order to better understand the role of the aforementioned factors in tumoral processes, we outline the latest models of intratumoral heterogeneity in glioblastoma multiforme. Based on the most recent reports, we discuss the problems of multi-drug therapy in treating glioblastoma multiforme.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland.,Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biała, 43-309 Bielsko-Biała, Poland
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Ireneusz Kojder
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland.,Department of Neurosurgery, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Dariusz Jeżewski
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland.,Department of Neurosurgery, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Marta Goschorska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Agnieszka Łukomska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| |
Collapse
|
16
|
Maschauer S, Prante O. Radiopharmaceuticals for imaging and endoradiotherapy of neurotensin receptor-positive tumors. J Labelled Comp Radiopharm 2018; 61:309-325. [DOI: 10.1002/jlcr.3581] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/13/2017] [Accepted: 10/24/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Simone Maschauer
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine; Friedrich Alexander University Erlangen-Nürnberg (FAU); Erlangen Germany
| | - Olaf Prante
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine; Friedrich Alexander University Erlangen-Nürnberg (FAU); Erlangen Germany
| |
Collapse
|
17
|
Neurotensin receptor 1 facilitates intracellular and transepithelial delivery of macromolecules. Eur J Pharm Biopharm 2017; 119:300-309. [DOI: 10.1016/j.ejpb.2017.06.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/02/2017] [Accepted: 06/28/2017] [Indexed: 01/11/2023]
|
18
|
New Insights in the Design of Bioactive Peptides and Chelating Agents for Imaging and Therapy in Oncology. Molecules 2017; 22:molecules22081282. [PMID: 28767081 PMCID: PMC6152110 DOI: 10.3390/molecules22081282] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/25/2017] [Indexed: 11/16/2022] Open
Abstract
Many synthetic peptides have been developed for diagnosis and therapy of human cancers based on their ability to target specific receptors on cancer cell surface or to penetrate the cell membrane. Chemical modifications of amino acid chains have significantly improved the biological activity, the stability and efficacy of peptide analogues currently employed as anticancer drugs or as molecular imaging tracers. The stability of somatostatin, integrins and bombesin analogues in the human body have been significantly increased by cyclization and/or insertion of non-natural amino acids in the peptide sequences. Moreover, the overall pharmacokinetic properties of such analogues and others (including cholecystokinin, vasoactive intestinal peptide and neurotensin analogues) have been improved by PEGylation and glycosylation. Furthermore, conjugation of those peptide analogues to new linkers and bifunctional chelators (such as AAZTA, TETA, TRAP, NOPO etc.), produced radiolabeled moieties with increased half life and higher binding affinity to the cognate receptors. This review describes the most important and recent chemical modifications introduced in the amino acid sequences as well as linkers and new bifunctional chelators which have significantly improved the specificity and sensitivity of peptides used in oncologic diagnosis and therapy.
Collapse
|
19
|
Jia Y, Zhang W, Fan W, Brusnahan S, Garrison J. Investigation of the Biological Impact of Charge Distribution on a NTR1-Targeted Peptide. Bioconjug Chem 2016; 27:2658-2668. [PMID: 27661393 DOI: 10.1021/acs.bioconjchem.6b00418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The neurotensin receptor 1 (NTR1) has been shown to be a promising target, due to its increased level of expression relative to normal tissue, for pancreatic and colon cancers. This has prompted the development of a variety of NTR1-targeted radiopharmaceuticals, based on the neurotensin (NT) peptide, for diagnostic and radiotherapeutic applications. A major obstacle for the clinical translation of NTR1-targeted radiotherapeutics would likely be nephrotoxicity due to the high levels of kidney retention. It is well-known that for many peptide-based agents, renal uptake is influenced by the overall molecular charge. Herein, we investigated the effect of charge distribution on receptor binding and kidney retention. Using the [(N-α-Me)Arg8,Dmt11,Tle12]NT(6-13) targeting vector, three peptides (177Lu-K2, 177Lu-K4, and 177Lu-K6), with the Lys moved closer (K6) or further away (K2) from the pharmacophore, were synthesized. In vitro competitive binding, internalization and efflux, and confocal microscopy studies were conducted using the NTR1-positive HT-29, human colon cancer cell line. The 177/natLu-K6 demonstrated the highest binding affinity (21.8 ± 1.2 nM) and the highest level of internalization (4.06% ± 0.20% of the total added amount). In vivo biodistribution, autoradiography, and metabolic studies of 177Lu-radiolabeled K2, K4, and K6 were examined using CF-1 mice. 177Lu-K4 and 177Lu-K6 gave the highest levels of in vivo uptake in NTR1-positive tissues, whereas 177Lu-K2 yielded nearly 2-fold higher renal uptake relative to the other radioconjugates. In conclusion, the position of the Lys (positively charged amino acid) influences the receptor binding, internalization, in vivo NTR1-targeting efficacy, and kidney retention profile of the radioconjugates. In addition, we have found that hydrophobicity likely play a role in the unique biodistribution profiles of these agents.
Collapse
Affiliation(s)
- Yinnong Jia
- Department of Pharmaceutical Sciences, College of Pharmacy, ‡Center for Drug Delivery and Nanomedicine, §Department of Biochemistry and Molecular Biology, College of Medicine, and ∥Eppley Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 985830, United States
| | - Wenting Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, ‡Center for Drug Delivery and Nanomedicine, §Department of Biochemistry and Molecular Biology, College of Medicine, and ∥Eppley Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 985830, United States
| | - Wei Fan
- Department of Pharmaceutical Sciences, College of Pharmacy, ‡Center for Drug Delivery and Nanomedicine, §Department of Biochemistry and Molecular Biology, College of Medicine, and ∥Eppley Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 985830, United States
| | - Susan Brusnahan
- Department of Pharmaceutical Sciences, College of Pharmacy, ‡Center for Drug Delivery and Nanomedicine, §Department of Biochemistry and Molecular Biology, College of Medicine, and ∥Eppley Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 985830, United States
| | - Jered Garrison
- Department of Pharmaceutical Sciences, College of Pharmacy, ‡Center for Drug Delivery and Nanomedicine, §Department of Biochemistry and Molecular Biology, College of Medicine, and ∥Eppley Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 985830, United States
| |
Collapse
|
20
|
Maschauer S, Einsiedel J, Hübner H, Gmeiner P, Prante O. 18F- and 68Ga-Labeled Neurotensin Peptides for PET Imaging of Neurotensin Receptor 1. J Med Chem 2016; 59:6480-92. [DOI: 10.1021/acs.jmedchem.6b00675] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Simone Maschauer
- Department
of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander University (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Jürgen Einsiedel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich-Alexander University (FAU), Schuhstraße 19, 91052 Erlangen, Germany
| | - Harald Hübner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich-Alexander University (FAU), Schuhstraße 19, 91052 Erlangen, Germany
| | - Peter Gmeiner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich-Alexander University (FAU), Schuhstraße 19, 91052 Erlangen, Germany
| | - Olaf Prante
- Department
of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander University (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| |
Collapse
|
21
|
Gupta SL, Dhiman V, Jayasekharan T, Sahoo NK. Analysis of argentinated peptide complexes using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry: Peptide = oxytocin, arg(8) -vasopressin, bradykinin, bombesin, somatostatin, neurotensin. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2016; 30:1313-1322. [PMID: 27173113 DOI: 10.1002/rcm.7562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/11/2016] [Accepted: 03/12/2016] [Indexed: 06/05/2023]
Abstract
RATIONALE The increased use of silver nanoparticles (AgNPs) for various biological applications, and over-expression of various peptide receptors in different tumors/cancer cells, necessitate the need for dedicated investigations on the intrinsic binding ability of Ag with various biologically important peptides for better understanding of AgNPs-peptide interactions and for the future development of contrasting agents as well as drugs for imaging/biomedical applications. METHODS The [M+(Ag)n ](+) complexes are prepared and characterized using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOFMS). RESULTS Silver complexes of the peptides [M+(Ag)n ](+) , where M = oxytocin, arg(8) -vasopressin, bradykinin, bombesin, somatostatin, and neurotensin, have been investigated for their intrinsic Ag(+) -binding ability. Unusual binding of up to seven Ag(+) with these small peptides is observed. The mass spectra show n = 1-5 for bombesin and somatostatin, n = 1-6 for bradykinin and arg(8) -vasopressin, and n = 1-7 for oxytocin and neurotensin. In addition, oxytocin and arg(8) -vasopressin show the formation of dimers and their complexes [M2 +(Ag)n ](+) with n = 1-8 and n = 1-5, respectively. The possible amino acid residues responsible for Ag(+) binding in each peptide have been identified on the basis of density functional theory (DFT)-calculated binding energy values of Ag(+) towards individual amino acids. CONCLUSIONS Mass spectrometric evidence indicates that the peptides, viz., oxytocin, arg(8) -vasopressin, bradykinin, bombesin, somatostatin, and neurotensin, show greater affinity for Ag(+) . Hence, they may be used as carriers for AgNPs in targeted drug delivery as well as an alternative for iodinated contrasting agents in dual energy X-ray imaging techniques. Radio-labeled Ag with these peptides can also be used in radio-pharmaceuticals for diagnostic and therapeutic applications. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Shyam L Gupta
- Atomic & Molecular Physics Division, Physics Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India
| | - Vikas Dhiman
- Atomic & Molecular Physics Division, Physics Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India
| | - T Jayasekharan
- Atomic & Molecular Physics Division, Physics Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India
| | - N K Sahoo
- Atomic & Molecular Physics Division, Physics Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India
| |
Collapse
|