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
|
Zhang T, Ma X, Xu M, Cai J, Cai J, Cao Y, Zhang Z, Ji X, He J, Cabrera GOF, Wu X, Zhao W, Wu Z, Xie J, Li Z. Chelator boosted tumor-retention and pharmacokinetic properties: development of 64Cu labeled radiopharmaceuticals targeting neurotensin receptor. Eur J Nucl Med Mol Imaging 2024; 51:3322-3333. [PMID: 38771516 PMCID: PMC11368631 DOI: 10.1007/s00259-024-06754-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/05/2024] [Indexed: 05/22/2024]
Abstract
PURPOSE Accumulating evidence suggests that neurotensin (NTS) and neurotensin receptors (NTSRs) play key roles in lung cancer progression by triggering multiple oncogenic signaling pathways. This study aims to develop Cu-labeled neurotensin receptor 1 (NTSR1)-targeting agents with the potential for both imaging and therapeutic applications. METHOD A series of neurotensin receptor antagonists (NRAs) with variable propylamine (PA) linker length and different chelators were synthesized, including [64Cu]Cu-CB-TE2A-iPA-NRA ([64Cu]Cu-4a-c, i = 1, 2, 3), [64Cu]Cu-NOTA-2PA-NRA ([64Cu]Cu-4d), [64Cu]Cu-DOTA-2PA-NRA ([64Cu]Cu-4e, also known as [64Cu]Cu-3BP-227), and [64Cu]Cu-DOTA-VS-2PA-NRA ([64Cu]Cu-4f). The series of small animal PET/CT were conducted in H1299 lung cancer model. The expression profile of NTSR1 was also confirmed by IHC using patient tissue samples. RESULTS For most of the compounds studied, PET/CT showed prominent tumor uptake and high tumor-to-background contrast, but the tumor retention was strongly influenced by the chelators used. For previously reported 4e, [64Cu]Cu-labeled derivative showed initial high tumor uptake accompanied by rapid tumor washout at 24 h. The newly developed [64Cu]Cu-4d and [64Cu]Cu-4f demonstrated good tumor uptake and tumor-to-background contrast at early time points, but were less promising in tumor retention. In contrast, our lead compound [64Cu]Cu-4b demonstrated 9.57 ± 1.35, 9.44 ± 2.38 and 9.72 ± 4.89%ID/g tumor uptake at 4, 24, and 48 h p.i., respectively. Moderate liver uptake (11.97 ± 3.85, 9.80 ± 3.63, and 7.72 ± 4.68%ID/g at 4, 24, and 48 h p.i.) was observed with low uptake in most other organs. The PA linker was found to have a significant effect on drug distribution. Compared to [64Cu]Cu-4b, [64Cu]Cu-4a had a lower background, including a greatly reduced liver uptake, while the tumor uptake was only moderately reduced. Meanwhile, [64Cu]Cu-4c showed increased uptake in both the tumor and the liver. The clinical relevance of NTSR1 was also demonstrated by the elevated tumor expression in patient tissue samples. CONCLUSIONS Through the side-by-side comparison, [64Cu]Cu-4b was identified as the lead agent for further evaluation based on its high and sustained tumor uptake and moderate liver uptake. It can not only be used to efficiently detect NTSR1 expression in lung cancer (for diagnosis, patient screening, and treatment monitoring), but also has the great potential to treat NTSR-positive lesions once chelating to the beta emitter 67Cu.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA.
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Transformation Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Xinrui Ma
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, Raleigh, NC, North Carolina State University, NC 27599, USA
| | - Muyun Xu
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Jinghua Cai
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Jianhua Cai
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zhihao Zhang
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Transformation Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xin Ji
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Transformation Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - German Oscar Fonseca Cabrera
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Xuedan Wu
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Weiling Zhao
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Zhanhong Wu
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Jin Xie
- Department of Chemistry, University of Georgia, 302 East Campus Road, Athens, GA, 30602, USA.
| | - Zibo Li
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA.
| |
Collapse
|
3
|
Abstract
Introduction: Neurotensin is a gut-brain peptide hormone, a 13 amino acid neuropeptide found in the central nervous system and in the GI tract. The neurotensinergic system is implicated in various physiological and pathological processes related to neuropsychiatric and metabolic machineries, cancer growth, food, and drug intake. NT mediates its functions through its two G protein-coupled receptors: neurotensin receptor 1 (NTS1/NTSR1) and neurotensin receptor 2 (NTS2/NTSR2). Over the past decade, the role of NTS3/NTSR3/sortilin has also gained importance in human pathologies. Several approaches have appeared dealing with the discovery of compounds able to modulate the functions of this neuropeptide through its receptors for therapeutic gain.Areas covered: The article provides an overview of over four decades of research and details the drug discovery approaches and patented strategies targeting NTSR in the past decade.Expert opinion: Neurotensin is an important neurotransmitter that enables crosstalk with various neurotransmitter and neuroendocrine systems. While significant efforts have been made that have led to selective agonists and antagonists with promising in vitro and in vivo activities, the therapeutic potential of compounds targeting the neurotensinergic system is still to be fully harnessed for successful clinical translation of compounds for the treatment of several pathologies.
Collapse
Affiliation(s)
- Malliga R Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
4
|
Neurotensins and their therapeutic potential: research field study. Future Med Chem 2020; 12:1779-1803. [PMID: 33032465 DOI: 10.4155/fmc-2020-0124] [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: 11/17/2022] Open
Abstract
The natural tridecapeptide neurotensin has been emerged as a promising therapeutic scaffold for the treatment of neurological diseases and cancer. In this work, we aimed to identify the top 100 most cited original research papers as well as recent key studies related to neurotensins. The Web of Science Core Collection database was searched and the retrieved research articles were analyzed by using the VOSviewer software. The most cited original articles were published between 1973 and 2013. The top-cited article was by Carraway and Leeman reporting the discovery of neurotensin in 1973. The highly cited terms were associated with hypotension and angiotensin-converting-enzyme. The conducted analysis reveals the therapeutic potentials of neurotensin, and further impactful research toward its clinical development is warrantied.
Collapse
|
5
|
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]
|
6
|
Rangger C, Haubner R. Radiolabelled Peptides for Positron Emission Tomography and Endoradiotherapy in Oncology. Pharmaceuticals (Basel) 2020; 13:E22. [PMID: 32019275 PMCID: PMC7169460 DOI: 10.3390/ph13020022] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
This review deals with the development of peptide-based radiopharmaceuticals for the use with positron emission tomography and peptide receptor radiotherapy. It discusses the pros and cons of this class of radiopharmaceuticals as well as the different labelling strategies, and summarises approaches to optimise metabolic stability. Additionally, it presents different target structures and addresses corresponding tracers, which are already used in clinical routine or are being investigated in clinical trials.
Collapse
Affiliation(s)
| | - Roland Haubner
- Department of Nuclear Medicine, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria;
| |
Collapse
|
7
|
Abstract
Single photon emission computed tomography (SPECT) is the state-of-the-art imaging modality in nuclear medicine despite the fact that only a few new SPECT tracers have become available in the past 20 years. Critical for the future success of SPECT is the design of new and specific tracers for the detection, localization, and staging of a disease and for monitoring therapy. The utility of SPECT imaging to address oncologic questions is dependent on radiotracers that ideally exhibit excellent tissue penetration, high affinity to the tumor-associated target structure, specific uptake and retention in the malignant lesions, and rapid clearance from non-targeted tissues and organs. In general, a target-specific SPECT radiopharmaceutical can be divided into two main parts: a targeting biomolecule (e.g., peptide, antibody fragment) and a γ-radiation-emitting radionuclide (e.g., 99mTc, 123I). If radiometals are used as the radiation source, a bifunctional chelator is needed to link the radioisotope to the targeting entity. In a rational SPECT tracer design, these single components have to be critically evaluated in order to achieve a balance among the demands for adequate target binding, and a rapid clearance of the radiotracer. The focus of this chapter is to depict recent developments of tumor-targeted SPECT radiotracers for imaging of cancer diseases. Possibilities for optimization of tracer design and potential causes for design failure are discussed and highlighted with selected examples.
Collapse
|
8
|
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
|
9
|
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
|
10
|
Deng H, Wang H, Zhang H, Wang M, Giglio B, Ma X, Jiang G, Yuan H, Wu Z, Li Z. Imaging Neurotensin Receptor in Prostate Cancer With 64Cu-Labeled Neurotensin Analogs. Mol Imaging 2018; 16:1536012117711369. [PMID: 28849698 PMCID: PMC6081756 DOI: 10.1177/1536012117711369] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Neurotensin receptor 1 (NTR-1) is expressed and activated in prostate cancer cells. In this study, we explore the NTR expression in normal mouse tissues and study the positron emission tomography (PET) imaging of NTR in prostate cancer models. MATERIALS AND METHODS Three 64Cu chelators (1, 4, 7, 10-tetraazacyclododecane-1, 4, 7, 10-tetraacetic acid [DOTA], 1,4,7-triazacyclononane-N,N',N″-triacetic acid [NOTA], or AmBaSar) were conjugated to an NT analog. Neurotensin receptor binding affinity was evaluated using cell binding assay. The imaging profile of radiolabeled probes was compared in well-established NTR+ HT-29 tumor model. Stability of the probes was tested. The selected agents were further evaluated in human prostate cancer PC3 xenografts. RESULTS All 3 NT conjugates retained the majority of NTR binding affinity. In HT-29 tumor, all agents demonstrated prominent tumor uptake. Although comparable stability was observed, 64Cu-NOTA-NT and 64Cu-AmBaSar-NT demonstrated improved tumor to background contrast compared with 64Cu-DOTA-NT. Positron emission tomography/computed tomography imaging of the NTR expression in PC-3 xenografts showed high tumor uptake of the probes, correlating with the in vitro Western blot results. Blocking experiments further confirmed receptor specificity. CONCLUSIONS Our results demonstrated that 64Cu-labeled neurotensin analogs are promising imaging agents for NTR-positive tumors. These agents may help us identify NTR-positive lesions and predict which patients and individual tumors are likely to respond to novel interventions targeting NTR-1.
Collapse
Affiliation(s)
- Huaifu Deng
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,2 PET/CT Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui Wang
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - He Zhang
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,3 Department of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Mengzhe Wang
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ben Giglio
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiaofen Ma
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,4 Department of Medical Imaging, Provincial People's Hospital, Guangzhou, China
| | - Guihua Jiang
- 4 Department of Medical Imaging, Provincial People's Hospital, Guangzhou, China
| | - Hong Yuan
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhanhong Wu
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zibo Li
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
11
|
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
|
12
|
Emrarian I, Sadeghzadeh N, Abedi SM, Abediankenari S. New neurotensin analogue radiolabeled by 99m-technetium as a potential agent for tumor identification. Chem Biol Drug Des 2017; 91:304-313. [DOI: 10.1111/cbdd.13082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/24/2017] [Accepted: 07/27/2017] [Indexed: 01/20/2023]
Affiliation(s)
- Iman Emrarian
- Department of Radiopharmacy; Faculty of Pharmacy; Mazandaran University of Medical Sciences; Sari Iran
| | - Nourollah Sadeghzadeh
- Department of Radiopharmacy; Faculty of Pharmacy; Mazandaran University of Medical Sciences; Sari Iran
| | - Seyed Mohammad Abedi
- Department of Radiology; Faculty of Medicine; Mazandaran University of Medical Sciences; Sari Iran
| | - Saeid Abediankenari
- Immunogenetics Research Center; Mazandaran University of Medical Sciences; Sari Iran
| |
Collapse
|
13
|
Charron CL, Hickey JL, Nsiama TK, Cruickshank DR, Turnbull WL, Luyt LG. Molecular imaging probes derived from natural peptides. Nat Prod Rep 2017; 33:761-800. [PMID: 26911790 DOI: 10.1039/c5np00083a] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Covering: up to the end of 2015.Peptides are naturally occurring compounds that play an important role in all living systems and are responsible for a range of essential functions. Peptide receptors have been implicated in disease states such as oncology, metabolic disorders and cardiovascular disease. Therefore, natural peptides have been exploited as diagnostic and therapeutic agents due to the unique target specificity for their endogenous receptors. This review discusses a variety of natural peptides highlighting their discovery, endogenous receptors, as well as their derivatization to create molecular imaging agents, with an emphasis on the design of radiolabelled peptides. This review also highlights methods for discovering new and novel peptides when knowledge of specific targets and endogenous ligands are not available.
Collapse
Affiliation(s)
- C L Charron
- Department of Chemistry, The University of Western Ontario, London, Canada.
| | - J L Hickey
- Department of Chemistry, The University of Western Ontario, London, Canada.
| | - T K Nsiama
- London Regional Cancer Program, Lawson Health Research Institute, London, Canada
| | - D R Cruickshank
- Department of Chemistry, The University of Western Ontario, London, Canada.
| | - W L Turnbull
- Department of Chemistry, The University of Western Ontario, London, Canada.
| | - L G Luyt
- Department of Chemistry, The University of Western Ontario, London, Canada. and Departments of Oncology and Medical Imaging, The University of Western Ontario, London, Canada and London Regional Cancer Program, Lawson Health Research Institute, London, Canada
| |
Collapse
|
14
|
Schulz J, Rohracker M, Stiebler M, Goldschmidt J, Stöber F, Noriega M, Pethe A, Lukas M, Osterkamp F, Reineke U, Höhne A, Smerling C, Amthauer H. Proof of Therapeutic Efficacy of a 177Lu-Labeled Neurotensin Receptor 1 Antagonist in a Colon Carcinoma Xenograft Model. J Nucl Med 2017; 58:936-941. [PMID: 28254866 DOI: 10.2967/jnumed.116.185140] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/05/2017] [Indexed: 11/16/2022] Open
Abstract
Increased expression of neurotensin receptor 1 (NTR1) has been shown in a large number of tumor entities such as pancreatic or colon carcinoma. Hence, this receptor is a promising target for diagnostic imaging and radioligand therapy. Using the favorable biodistribution data of the NTR1-targeting agent 111In-3BP-227, we investigated the therapeutic effect of its 177Lu-labeled analog on the tumor growth of NTR1-positive HT29 colon carcinoma xenografts. Methods: 3BP-227 was labeled with 177Lu. To assess its biodistribution properties, SPECT and CT scans of HT29-xenografted nude mice injected with 177Lu-3BP-227 were acquired, and ex vivo tissue activity was determined. To evaluate therapeutic efficacy, 2 groups of mice received the radiopharmaceutical in a median dose of either 165 MBq (129-232 MBq, n = 10) or 110 MBq (82-116 MBq, n = 10), whereas control mice were injected with vehicle (n = 10). Tumor sizes and body weights were monitored for up to 49 d. Renal function and histologic morphology were evaluated. Results: Whole-body SPECT/CT images allowed clear tumor visualization with low background activity and high tumor-to-kidney and -liver ratios. Ex vivo biodistribution data confirmed high and persistent uptake of 177Lu-3BP-227 in HT29 tumors (19.0 ± 3.6 vs. 2.7 ± 1.6 percentage injected dose per gram at 3 and 69 h after injection, respectively). The application of 177Lu-3BP-227 resulted in a distinct delay of tumor growth. Median tumor doubling time for controls was 5.5 d (interquartile range [IQR], 2.8-7.0), compared with 17.5 d (IQR, 5.5-22.5 d) for the 110-MBq and 41.0 d (IQR, 27.5-55.0) for the 165-MBg group. Compared with controls, median relative tumor volume at day 23 after injection was reduced by 55% (P = 0.034) in the 110-MBq and by 88% (P < 0.01) in the 165-MBq group. Renal histology and clinical chemistry results did not differ between radiotherapy groups and controls, suggesting absence of therapy-induced acute renal damage. Conclusion: These data demonstrate that the novel NTR1-targeting theranostic agent 3BP-227 is an effective and promising candidate for radioligand therapy, with a favorable preliminary safety profile and high potential for clinical translation.
Collapse
Affiliation(s)
- Jörg Schulz
- Klinik für Radiologie und Nuklearmedizin, Otto-von-Guericke Universität, Magdeburg, Germany
| | - Martin Rohracker
- Klinik für Radiologie und Nuklearmedizin, Otto-von-Guericke Universität, Magdeburg, Germany
| | - Marvin Stiebler
- Klinik für Radiologie und Nuklearmedizin, Otto-von-Guericke Universität, Magdeburg, Germany
| | | | - Franziska Stöber
- Klinik für Radiologie und Nuklearmedizin, Otto-von-Guericke Universität, Magdeburg, Germany.,Leibniz-Institut für Neurobiologie, Magdeburg, Germany
| | - Mercedes Noriega
- Institut für Pathologie, Universitätsklinik Hamburg-Eppendorf, Hamburg, Germany
| | - Anette Pethe
- Klinik für Radiologie und Nuklearmedizin, Otto-von-Guericke Universität, Magdeburg, Germany
| | - Mathias Lukas
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Siemens Healthcare GmbH, Erlangen, Germany; and
| | | | | | | | | | - Holger Amthauer
- Klinik für Radiologie und Nuklearmedizin, Otto-von-Guericke Universität, Magdeburg, Germany .,Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
15
|
A Review of the Role of Neurotensin and Its Receptors in Colorectal Cancer. Gastroenterol Res Pract 2017; 2017:6456257. [PMID: 28316623 PMCID: PMC5339424 DOI: 10.1155/2017/6456257] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/24/2017] [Indexed: 12/25/2022] Open
Abstract
Neurotensin (NTS) is a physiologically occurring hormone which affects the function of the gastrointestinal (GI) tract. In recent years, NTS, acting through its cellular receptors (NTSR), has been implicated in the carcinogenesis of several cancers. In colorectal cancer (CRC), a significant body of evidence, from in vitro and in vivo studies, is available which elucidates the molecular biology of NTS/NTSR signalling and the resultant growth of CRC cells. There is growing clinical data from human studies which corroborate the role NTS/NTSR plays in the development of human CRC. Furthermore, blockade and modulation of the NTS/NTSR signalling pathways appears to reduce CRC growth in cell cultures and animal studies. Lastly, NTS/NTSR also shows potential of being utilised as a diagnostic biomarker for cancers as well as targets for functional imaging. We summarise the existing evidence and understanding of the role of NTS and its receptors in CRC.
Collapse
|
16
|
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
|
17
|
Development of Drugs and Technology for Radiation Theragnosis. NUCLEAR ENGINEERING AND TECHNOLOGY 2016. [DOI: 10.1016/j.net.2016.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
18
|
Jia Y, Shi W, Zhou Z, Wagh NK, Fan W, Brusnahan SK, Garrison JC. Evaluation of DOTA-chelated neurotensin analogs with spacer-enhanced biological performance for neurotensin-receptor-1-positive tumor targeting. Nucl Med Biol 2015; 42:816-23. [PMID: 26302836 DOI: 10.1016/j.nucmedbio.2015.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/25/2015] [Accepted: 07/21/2015] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Neurotensin receptor 1 (NTR1) is overexpressed in many cancer types. Neurotensin (NT), a 13 amino acid peptide, is the native ligand for NTR1 and exhibits high (nM) affinity to the receptor. Many laboratories have been investigating the development of diagnostic and therapeutic radiopharmaceuticals for NTR1-positive cancers based on the NT peptide. To improve the biological performance for targeting NTR1, we proposed NT analogs with a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelation system and different lengths of spacers. METHODS We synthesized four NTR1-targeted conjugates with spacer lengths from 0 to 9 atoms (null (N0), β-Ala-OH (N1), 5-Ava-OH (N2), and 8-Aoc-OH (N3)) between the DOTA and the pharmacophore. In vitro competitive binding, internalization and efflux studies were performed on all four NT analogs. Based on these findings, metabolism studies were carried out on our best performing conjugate, (177)Lu-N1. Lastly, in vivo biodistribution and SPECT/CT imaging studies were performed using (177)Lu-N1 in an HT-29 xenograft mouse model. RESULTS As shown in the competitive binding assays, the NT analogs with different spacers (N1, N2 and N3) exhibited lower IC50 values than the NT analog without a spacer (N0). Furthermore, N1 revealed higher retention in HT-29 cells with more rapid internalization and slower efflux than the other NT analogs. In vivo biodistribution and SPECT/CT imaging studies of (177)Lu-N1 demonstrated excellent accumulation (3.1 ± 0.4%ID/g) in the NTR1-positive tumors at 4h post-administration. CONCLUSIONS The DOTA chelation system demonstrated some modest steric inhibition of the pharmacophore. However, the insertion of a 4-atom hydrocarbon spacer group restored optimal binding affinity of the analog. The in vivo assays indicated that (177)Lu-N1 could be used for imaging and radiotherapy of NTR1-positive tumors.
Collapse
Affiliation(s)
- Yinnong Jia
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA, 68198-5830
| | - Wen Shi
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA, 68198-5830
| | - Zhengyuan Zhou
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA, 68198-5830
| | - Nilesh K Wagh
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA, 68198-5830
| | - Wei Fan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA, 68198-5830
| | - Susan K Brusnahan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA, 68198-5830
| | - Jered C Garrison
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE, USA, 68198-5830.
| |
Collapse
|
19
|
Deng H, Wang H, Wang M, Li Z, Wu Z. Synthesis and Evaluation of 64Cu-DOTA-NT-Cy5.5 as a Dual-Modality PET/Fluorescence Probe to Image Neurotensin Receptor-Positive Tumor. Mol Pharm 2015; 12:3054-61. [PMID: 26162008 DOI: 10.1021/acs.molpharmaceut.5b00325] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Overexpression of neurotensin receptors (NTRs) has been suggested to play important roles in the growth and survival of a variety of tumor types. The aim of this study is to develop a dual-modality probe (64Cu -DOTA-NT-Cy5.5) for imaging NTR1 expression in vivo with both positron emission tomography (PET) and fluorescence. In this approach, the thiol group and N terminal amino group of neurotensin analogue (Cys-NT) were chemically modified with Cy5.5 dye and DOTA chelator, respectively. After radiolabeling with 64Cu, the resulting probe (64Cu-DOTA-NT-Cy5.5) was evaluated in NTR1 positive HT-29 tumor model. Small animal PET quantification analysis demonstrated that the tumor uptake was 1.91±0.22 and 1.79±0.16%ID/g at 1 and 4 h postinjection (p.i.), respectively. The tumor-to-muscle ratio was 17.44±3.25 at 4 h p.i. based on biodistribution. Receptor specificity was confirmed by the successful blocking experiment at 4 h p.i. (0.42±0.05%ID/g). In parallel with PET experiment, fluorescence imaging was also performed, which demonstrated prominent tumor uptake in HT-29 model. As a proof of concept, an imaging guided surgery was performed to the fluorescent moiety of this probe and could provide potential surgery guidance for NTR positive patients. In summary, our results clearly indicated that the dual-modality probe, 64Cu-DOTA-NT-Cy5.5, could serve as a promising agent to image NTR positive tumors in vivo.
Collapse
Affiliation(s)
- Huaifu Deng
- †Biomedical Research Imaging Center and Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States.,‡PET/CT Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Hui Wang
- †Biomedical Research Imaging Center and Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mengzhe Wang
- †Biomedical Research Imaging Center and Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Zibo Li
- †Biomedical Research Imaging Center and Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Zhanhong Wu
- †Biomedical Research Imaging Center and Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
20
|
Körner M, Waser B, Strobel O, Büchler M, Reubi JC. Neurotensin receptors in pancreatic ductal carcinomas. EJNMMI Res 2015; 5:17. [PMID: 25859423 PMCID: PMC4388205 DOI: 10.1186/s13550-015-0094-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/26/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The frequent expression of neurotensin receptors (NT-R) in primaries of pancreatic ductal carcinomas has triggered the development of radioactive neurotensin analogs for possible in vivo targeting of these tumors. However, the complete lack of information regarding NT-R in liver metastases of pancreatic cancer and pancreatic intraepithelial neoplasia (PanIN) makes an in vitro study of NT-R in these tissues indispensable. METHODS Using in vitro receptor autoradiography with (125)I-[Tyr(3)]-neurotensin, NT-R were investigated in 18 primaries and 23 liver metastases of pancreatic ductal carcinomas as well as in 19 PanIN lesions. RESULTS We report here that 13 of 18 ductal carcinoma primaries and 14 of 23 liver metastases expressed NT-R. Moreover, none of the six PanIN 1B cases expressed NT-R, while two of six PanIN 2 and five of seven PanIN 3 expressed NT-R. Binding was fully displaced by the type 1 NT-R-selective antagonist SR48692, indicating that the NT-R in the tumors are of the type 1 NT-R subtype. CONCLUSIONS These in vitro data extend the currently available information on NT-R in invasive and non-invasive pancreatic ductal tumors. They suggest that type 1 NT-R may be a novel, specific marker of PanIN of higher degree. The high expression of NT-R in primaries and metastases of invasive cancer strongly support the need to develop radioactive neurotensin analogs for the diagnosis and therapy of this tumor type.
Collapse
Affiliation(s)
- Meike Körner
- Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, PO Box 62, Murtenstrasse 31, CH-3010 Berne, Switzerland
| | - Beatrice Waser
- Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, PO Box 62, Murtenstrasse 31, CH-3010 Berne, Switzerland
| | - Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Markus Büchler
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Jean Claude Reubi
- Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, PO Box 62, Murtenstrasse 31, CH-3010 Berne, Switzerland
| |
Collapse
|
21
|
Ye Y, Liu P, Wang Y, Li H, Wei F, Cheng Y, Han L, Yu J. Neurotensin, a Novel Messenger to Cross-Link Inflammation and Tumor Invasion via Epithelial-Mesenchymal Transition Pathway. Int Rev Immunol 2014; 35:340-350. [PMID: 25215420 DOI: 10.3109/08830185.2014.952412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Multiple cytokines and growth factors are critical for the prognosis of cancer which has been regarded as a worldwide health problem. Recently, neuropeptides, soluble factors regulating a series of functions in the central nervous system, have also been demonstrated to stimulate the proliferation and migration of tumor cells. Among these signaling peptides, the role of neurotensin (NTS) on malignancy procession has become a hot topic. The effects of NTS on tumor growth and its antiapoptosis role have already been identified. Subsequently, studies demonstrated the impact of NTS on the migration and invasion, but the molecular mechanisms involved are still unclear at present. Recently, some reports indicated that NTS could induce expression and secretion of interleukin-8 (IL-8) to promote local imflammatory response which might participate in epithelial-mesenchymal transition (EMT)-related tumor migration. In present review, we highlight the process of tumor EMT induced by NTS through stimulating IL-8 and the significance of NTS/IL-8 pathway in clinical application prospect.
Collapse
Affiliation(s)
- Yingnan Ye
- a Department of Immunology , Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy , Tianjin , P. R. China
| | - Pengpeng Liu
- b Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy , Tianjin , P. R. China
| | - Yue Wang
- a Department of Immunology , Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy , Tianjin , P. R. China
| | - Hui Li
- a Department of Immunology , Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy , Tianjin , P. R. China
| | - Feng Wei
- a Department of Immunology , Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy , Tianjin , P. R. China
| | - Yanan Cheng
- b Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy , Tianjin , P. R. China
| | - Lei Han
- b Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy , Tianjin , P. R. China
| | - Jinpu Yu
- a Department of Immunology , Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy , Tianjin , P. R. China.,b Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy , Tianjin , P. R. China.,c Biotherapy Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy , Tianjin , P. R. China
| |
Collapse
|
22
|
Morgat C, Mishra AK, Varshney R, Allard M, Fernandez P, Hindié E. Targeting neuropeptide receptors for cancer imaging and therapy: perspectives with bombesin, neurotensin, and neuropeptide-Y receptors. J Nucl Med 2014; 55:1650-7. [PMID: 25189338 DOI: 10.2967/jnumed.114.142000] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Receptors for some regulatory peptides are highly expressed in tumors. Selective radiolabeled peptides can bind with high affinity and specificity to these receptors and exhibit favorable pharmacologic and pharmacokinetic properties, making them suitable agents for imaging or targeted therapy. The success encountered with radiolabeled somatostatin analogs is probably the first of a long list, as multiple peptide receptors are now recognized as potential targets. This review focuses on 3 neuropeptide receptor systems (bombesin, neurotensin, and neuropeptide-Y) that offer high potential in the field of nuclear oncology. The underlying biology of these peptide/receptor systems, their physiologic and pathologic roles, and their differential distribution in normal and tumoral tissues are described with emphasis on breast, prostate, and lung cancers. Radiolabeled analogs that selectively target these receptors are highlighted.
Collapse
Affiliation(s)
- Clément Morgat
- CHU de Bordeaux, Service de Médecine Nucléaire, Bordeaux, France University of Bordeaux, INCIA, UMR 5287, Talence, France CNRS, INCIA, UMR 5287, Talence, France
| | - Anil Kumar Mishra
- University of Bordeaux, INCIA, UMR 5287, Talence, France CNRS, INCIA, UMR 5287, Talence, France Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, New Delhi, India; and
| | - Raunak Varshney
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, New Delhi, India; and
| | - Michèle Allard
- CHU de Bordeaux, Service de Médecine Nucléaire, Bordeaux, France University of Bordeaux, INCIA, UMR 5287, Talence, France EPHE, Bordeaux, France
| | - Philippe Fernandez
- CHU de Bordeaux, Service de Médecine Nucléaire, Bordeaux, France University of Bordeaux, INCIA, UMR 5287, Talence, France CNRS, INCIA, UMR 5287, Talence, France
| | - Elif Hindié
- CHU de Bordeaux, Service de Médecine Nucléaire, Bordeaux, France University of Bordeaux, INCIA, UMR 5287, Talence, France CNRS, INCIA, UMR 5287, Talence, France
| |
Collapse
|
23
|
Chanawanno K, Caporoso J, Kondeti V, Paruchuri S, Leeper TC, Herrick RS, Ziegler CJ. Facile solid phase peptide synthesis with a Re-lysine conjugate generated via a one-pot procedure. Dalton Trans 2014; 43:11452-5. [PMID: 24875597 DOI: 10.1039/c4dt01129e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We have synthesized a Re(CO)3-modified lysine via a one-pot Schiff base formation reaction that can be used in the solid phase peptide synthesis. To demonstrate its potential use, we have attached it to a neurotensin fragment and observed uptake into human umbilical vascular endothelial cells.
Collapse
Affiliation(s)
- Kullapa Chanawanno
- Department of Chemistry, University of Akron, Akron, OH 44325-3601, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Guo Z, Du S, Chen B, Sha Y, Qiu B, Jiang X, Wang S, Li X. A sandwich-type label-free electrochemiluminescence immunosensor for neurotensin based on sombrero model with graphene-hyaluronate-luminol composite. Electrochim Acta 2014. [DOI: 10.1016/j.electacta.2014.05.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
25
|
A bombesin-shepherdin radioconjugate designed for combined extra- and intracellular targeting. Pharmaceuticals (Basel) 2014; 7:662-75. [PMID: 24871806 PMCID: PMC4078514 DOI: 10.3390/ph7060662] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/17/2014] [Accepted: 05/20/2014] [Indexed: 12/28/2022] Open
Abstract
Radiolabeled peptides which target tumor-specific membrane structures of cancer cells represent a promising class of targeted radiopharmaceuticals for the diagnosis and therapy of cancer. A potential drawback of a number of reported radiopeptides is the rapid washout of a substantial fraction of the initially delivered radioactivity from cancer cells and tumors. This renders the initial targeting effort in part futile and results in a lower imaging quality and efficacy of the radiotracer than achievable. We are investigating the combination of internalizing radiopeptides with molecular entities specific for an intracellular target. By enabling intracellular interactions of the radioconjugate, we aim at reducing/decelerating the externalization of radioactivity from cancer cells. Using the “click-to-chelate” approach, the 99mTc-tricarbonyl core as a reporter probe for single-photon emission computed tomography (SPECT) was combined with the binding sequence of bombesin for extracellular targeting of the gastrin-releasing peptide receptor (GRP-r) and peptidic inhibitors of the cytosolic heat shock 90 protein (Hsp90) for intracellular targeting. Receptor-specific uptake of the multifunctional radioconjugate could be confirmed, however, the cellular washout of radioactivity was not improved. We assume that either endosomal trapping or lysosomal degradation of the radioconjugate is accountable for these observations.
Collapse
|
26
|
Wu Z, Li L, Liu S, Yakushijin F, Yakushijin K, Horne D, Conti PS, Li Z, Kandeel F, Shively JE. Facile Preparation of a Thiol-Reactive (18)F-Labeling Agent and Synthesis of (18)F-DEG-VS-NT for PET Imaging of a Neurotensin Receptor-Positive Tumor. J Nucl Med 2014; 55:1178-84. [PMID: 24854793 DOI: 10.2967/jnumed.114.137489] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/17/2014] [Indexed: 01/07/2023] Open
Abstract
UNLABELLED Accumulating evidence suggests that neurotensin receptors (NTRs) play key roles in cancer growth and survival. In this study, we developed a simple and efficient method to radiolabel neurotensin peptide with (18)F for NTR-targeted imaging. METHODS The thiol-reactive reagent (18)F-(2-(2-(2-fluoroethoxy)ethoxy)ethylsulfonyl)ethane ((18)F-DEG-VS) was facilely prepared through 1-step radiofluorination. After high-pressure liquid chromatography purification, (18)F-DEG-VS was incubated with the c(RGDyC) and c(RGDyK) peptide mixture to evaluate its specificity toward the reactive thiol. Thiolated neurotensin peptide was then labeled with (18)F using this novel synthon, and the resulting imaging probe was subjected to receptor-binding assay and small-animal PET studies in a murine xenograft model. The imaging results and metabolic stability of (18)F-DEG-VS-NT were compared with the thiol-specific maleimide derivative N-[2-(4-(18)F-fluorobenzamido)ethyl]maleimide-neurotensin ((18)F-FBEM-NT). RESULTS (18)F-DEG-VS was obtained in high labeling yield. The reaction of (19)F-DEG-VS was highly specific for thiols at neutral pH, whereas the lysine of c(RGDyK) reacted at a pH greater than 8.5. (18)F-DEG-VS-c(RGDyC) was the preferred product when both c(RGDyK) and c(RGDyC) were incubated together with (18)F-DEG-VS. Thiolated neurotensin peptide (Cys-NT) efficiently reacted with (18)F-DEG-VS, with a 95% labeling yield (decay-corrected). The radiochemical purity of the (18)F-DEG-VS-NT was greater than 98%, and the specific activity was about 19.2 ± 4.3 TBq/mmol. Noninvasive small-animal PET demonstrated that (18)F-DEG-VS-NT had an NTR-specific tumor uptake in subcutaneous HT-29 xenografts. The tumor-to-muscle, tumor-to-liver, and tumor-to-kidney ratios reached 30.65 ± 22.31, 11.86 ± 1.98, and 1.91 ± 0.43 at 2 h after injection, respectively, based on the biodistribution study. Receptor specificity was demonstrated by blocking experiment. Compared with (18)F-FBEM-NT, (18)F-DEG-VS-NT was synthesized with fewer steps and provided significantly improved imaging quality in vivo. CONCLUSION We have established a facile (18)F-labeling method for site-specific labeling of the Cys-NT. Using this method, we synthesized an NTR-targeted PET agent, which demonstrated high tumor-to-background contrast.
Collapse
Affiliation(s)
- Zhanhong Wu
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of the City of Hope, Duarte, California
| | - Lin Li
- Department of Immunology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Shuanglong Liu
- Department of Radiology, Molecular Imaging Center, University of Southern California, Los Angeles, California; and
| | - Fumiko Yakushijin
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Kenichi Yakushijin
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Peter S Conti
- Department of Radiology, Molecular Imaging Center, University of Southern California, Los Angeles, California; and
| | - Zibo Li
- Department of Radiology, Molecular Imaging Center, University of Southern California, Los Angeles, California; and
| | - Fouad Kandeel
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of the City of Hope, Duarte, California
| | - John E Shively
- Department of Immunology, Beckman Research Institute of the City of Hope, Duarte, California
| |
Collapse
|
27
|
Maschauer S, Ruckdeschel T, Tripal P, Haubner R, Einsiedel J, Hübner H, Gmeiner P, Kuwert T, Prante O. In vivo monitoring of the antiangiogenic effect of neurotensin receptor-mediated radiotherapy by small-animal positron emission tomography: a pilot study. Pharmaceuticals (Basel) 2014; 7:464-81. [PMID: 24743103 PMCID: PMC4014703 DOI: 10.3390/ph7040464] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 04/04/2014] [Accepted: 04/10/2014] [Indexed: 12/22/2022] Open
Abstract
The neurotensin receptor (NTS1) has emerged as an interesting target for molecular imaging and radiotherapy of NTS-positive tumors due to the overexpression in a range of tumors. The aim of this study was to develop a 177Lu-labeled NTS1 radioligand, its application for radiotherapy in a preclinical model and the imaging of therapy success by small-animal positron emission tomography (µPET) using [68Ga]DOTA-RGD as a specific tracer for imaging angiogenesis. The 177Lu-labeled peptide was subjected to studies on HT29-tumor-bearing nude mice in vivo, defining four groups of animals (single dose, two fractionated doses, four fractionated doses and sham-treated animals). Body weight and tumor diameters were determined three times per week. Up to day 28 after treatment, µPET studies were performed with [68Ga]DOTA-RGD. At days 7–10 after treatment with four fractionated doses of 11–14 MBq (each at days 0, 3, 6 and 10), the tumor growth was slightly decreased in comparison with untreated animals. Using a single high dose of 51 MBq, a significantly decreased tumor diameter of about 50% was observed with the beginning of treatment. Our preliminary PET imaging data suggested decreased tumor uptake values of [68Ga]DOTA-RGD in treated animals compared to controls at day 7 after treatment. This pilot study suggests that early PET imaging with [68Ga]DOTA-RGD in radiotherapy studies to monitor integrin expression could be a promising tool to predict therapy success in vivo. Further successive PET experiments are needed to confirm the significance and predictive value of RGD-PET for NTS-mediated radiotherapy.
Collapse
Affiliation(s)
- Simone Maschauer
- Department of Nuclear Medicine, Laboratory of Molecular Imaging and Radiochemistry, Friedrich Alexander University, Schwabachanlage 6, 91054 Erlangen, Germany.
| | - Tina Ruckdeschel
- Department of Nuclear Medicine, Laboratory of Molecular Imaging and Radiochemistry, Friedrich Alexander University, Schwabachanlage 6, 91054 Erlangen, Germany.
| | - Philipp Tripal
- Department of Nuclear Medicine, Laboratory of Molecular Imaging and Radiochemistry, Friedrich Alexander University, Schwabachanlage 6, 91054 Erlangen, Germany.
| | - Roland Haubner
- Department of Nuclear Medicine, Innsbruck Medical University, Anichstr. 35, 6020 Innsbruck, Austria.
| | - Jürgen Einsiedel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany.
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany.
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany.
| | - Torsten Kuwert
- Department of Nuclear Medicine, Laboratory of Molecular Imaging and Radiochemistry, Friedrich Alexander University, Schwabachanlage 6, 91054 Erlangen, Germany.
| | - Olaf Prante
- Department of Nuclear Medicine, Laboratory of Molecular Imaging and Radiochemistry, Friedrich Alexander University, Schwabachanlage 6, 91054 Erlangen, Germany.
| |
Collapse
|
28
|
Lang C, Maschauer S, Hübner H, Gmeiner P, Prante O. Synthesis and evaluation of a (18)F-labeled diarylpyrazole glycoconjugate for the imaging of NTS1-positive tumors. J Med Chem 2013; 56:9361-5. [PMID: 24160350 DOI: 10.1021/jm401491e] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Aiming to image NTS1 overexpressing tumors, the diarylpyrazole glycoconjugate 8, derived from the potent NTS1 antagonist SR142948A, was synthesized taking advantage of the palladium-catalyzed aminocarbonylation reaction. The glycoconjugate 8 displayed excellent affinity and selectivity toward NTS1. Radiosynthesis proceeded straightforwardly, obtaining [(18)F]8 with excellent stability and highly beneficial biodistribution in vivo as demonstrated by PET imaging in HT29 tumor-bearing nude mice. Thus, the tracer [(18)F]8 represents a highly promising candidate for PET imaging of NTS1-positive tumors.
Collapse
Affiliation(s)
- Christopher Lang
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | | | | | | | | |
Collapse
|
29
|
Ahrabi NZ, Erfani M, Parivar K, Beiki D, Jalilian AR. Preparation and evaluation of a new neurotensin analog labeled with 99mTc for targeted imaging of neurotensin receptor positive tumors. J Radioanal Nucl Chem 2013. [DOI: 10.1007/s10967-013-2795-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
30
|
Abstract
Single photon emission computed tomography (SPECT) is the state-of-the-art imaging modality in nuclear medicine despite the fact that only a few new SPECT tracers have become available in the past 20 years. Critical for the future success of SPECT is the design of new and specific tracers for the detection, localization, and staging of a disease and for monitoring therapy. The utility of SPECT imaging to address oncologic questions is dependent on radiotracers that ideally exhibit excellent tissue penetration, high affinity to the tumor-associated target structure, specific uptake and retention in the malignant lesions, and rapid clearance from non-targeted tissues and organs. In general, a target-specific SPECT radiopharmaceutical can be divided into two main parts: a targeting biomolecule (e.g. peptide, antibody fragment) and a γ-radiation emitting radionuclide (e.g. (99m)Tc, (123)I). If radiometals are used as the radiation source, a bifunctional chelator is needed to link the radioisotope to the targeting entity. In a rational SPECT tracer design these single components have to be critically evaluated in order to achieve a balance among the demands for adequate target binding, and a rapid clearance of the radiotracer. The focus of this chapter is to depict recent developments of tumor-targeted SPECT radiotracers for imaging of cancer diseases. Possibilities for optimization of tracer design and potential causes for design failure are discussed and highlighted with selected examples.
Collapse
Affiliation(s)
- Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI, Switzerland.
| | | |
Collapse
|
31
|
Held C, Plomer M, Hübner H, Meltretter J, Pischetsrieder M, Gmeiner P. Development of a Metabolically Stable Neurotensin Receptor 2 (NTS2) Ligand. ChemMedChem 2012; 8:75-81. [DOI: 10.1002/cmdc.201200376] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 09/21/2012] [Indexed: 12/18/2022]
|
32
|
Kluba CA, Bauman A, Valverde IE, Vomstein S, Mindt TL. Dual-targeting conjugates designed to improve the efficacy of radiolabeled peptides. Org Biomol Chem 2012; 10:7594-602. [PMID: 22898743 DOI: 10.1039/c2ob26127h] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Radiolabeled regulatory peptides are useful tools in nuclear medicine for the diagnosis (imaging) and therapy of cancer. The specificity of the peptides towards GPC receptors, which are overexpressed by cancer cells, and their favorable pharmacokinetic profile make them ideal vectors to transport conjugated radionuclides to tumors and metastases. However, after internalization of the radiopeptide into cancer cells and tumors, a rapid washout of a substantial fraction of the delivered radioactivity is often observed. This phenomenon may represent a limitation of radiopeptides for clinical applications. Here, we report the synthesis, radiolabeling, stability, and in vitro evaluation of a novel, dual-targeting peptide radioconjugate designed to enhance the cellular retention of radioactivity. The described trifunctional conjugate is comprised of a Tc-99m SPECT reporter probe, a cell membrane receptor-specific peptide, and a second targeting entity directed towards mitochondria. While the specificity of the first generation of dual-targeting conjugates towards its extracellular target was demonstrated, intracellular targeting could not be confirmed probably due to non-specific binding or hindered passage through the membrane of the organelle. The work presented describes a novel approach with potential to improve the efficacy of radiopharmaceuticals by enhancing the intracellular retention of radioactivity.
Collapse
Affiliation(s)
- Christiane A Kluba
- University of Basel Hospital, Department of Radiology and Nuclear Medicine, Division of Radiological Chemistry, Petersgraben 4, 4031 Basel, Switzerland
| | | | | | | | | |
Collapse
|
33
|
Morais GR, Paulo A, Santos I. Organometallic Complexes for SPECT Imaging and/or Radionuclide Therapy. Organometallics 2012. [DOI: 10.1021/om300501d] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Goreti Ribeiro Morais
- Unidade de Ciências
Quı́micas e Radiofarmacêuticas, Instituto
Tecnológico e Nuclear, Instituto Superior Técnico, Universidade Técnica de Lisboa, Estrada Nacional
10, 2686-953, Sacavém, Portugal
| | - António Paulo
- Unidade de Ciências
Quı́micas e Radiofarmacêuticas, Instituto
Tecnológico e Nuclear, Instituto Superior Técnico, Universidade Técnica de Lisboa, Estrada Nacional
10, 2686-953, Sacavém, Portugal
| | - Isabel Santos
- Unidade de Ciências
Quı́micas e Radiofarmacêuticas, Instituto
Tecnológico e Nuclear, Instituto Superior Técnico, Universidade Técnica de Lisboa, Estrada Nacional
10, 2686-953, Sacavém, Portugal
| |
Collapse
|
34
|
Fani M, Maecke HR, Okarvi SM. Radiolabeled peptides: valuable tools for the detection and treatment of cancer. Am J Cancer Res 2012; 2:481-501. [PMID: 22737187 PMCID: PMC3364555 DOI: 10.7150/thno.4024] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/31/2012] [Indexed: 12/17/2022] Open
Abstract
Human cancer cells overexpress many peptide receptors as molecular targets. Radiolabeled peptides that bind with high affinity and specificity to the receptors on tumor cells hold great potential for both diagnostic imaging and targeted radionuclide therapy. The advantage of solid-phase peptide synthesis, the availability of different chelating agents and prosthetic groups and bioconjugation techniques permit the facile preparation of a wide variety of peptide-based targeting molecules with diverse biological and tumor targeting properties. Some of these peptides, including somatostatin, bombesin, vasoactive intestinal peptide, gastrin, neurotensin, exendin and RGD are currently under investigation. It is anticipated that in the near future many of these peptides may find applications in nuclear oncology. This article presents recent developments in the field of small peptides, and their applications in the diagnosis and treatment of cancer.
Collapse
|
35
|
Wu Z, Martinez-Fong D, Trédaniel J, Forgez P. Neurotensin and its high affinity receptor 1 as a potential pharmacological target in cancer therapy. Front Endocrinol (Lausanne) 2012; 3:184. [PMID: 23335914 PMCID: PMC3547287 DOI: 10.3389/fendo.2012.00184] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 12/26/2012] [Indexed: 12/12/2022] Open
Abstract
Cancer is a worldwide health problem. Personalized treatment represents a future advancement for cancer treatment, in part due to the development of targeted therapeutic drugs. These molecules are expected to be more effective than current treatments and less harmful to normal cells. The discovery and validation of new targets are the foundation and the source of these new therapies. The neurotensinergic system has been shown to enhance cancer progression in various cancers such as pancreatic, prostate, lung, breast, and colon cancer. It also triggers multiple oncogenic signaling pathways, such as the PKC/ERK and AKT pathways. In this review, we discuss the contribution of the neurotensinergic system to cancer progression, as well as the regulation and mechanisms of the system in order to highlight its potential as a therapeutic target, and its prospect for its use as a treatment in certain cancers.
Collapse
Affiliation(s)
- Zherui Wu
- INSERM-UPMC UMR_S938, Hôpital Saint-AntoineParis, France
| | - Daniel Martinez-Fong
- Departamento de Fisiologïa, Biofïsica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico NacionalMexico City, Mexico
| | - Jean Trédaniel
- INSERM-UPMC UMR_S938, Hôpital Saint-AntoineParis, France
- Unité de Cancérologie Thoracique, Groupe Hospitalier Paris Saint-Joseph/Université Paris DescartesParis, France
| | - Patricia Forgez
- INSERM-UPMC UMR_S938, Hôpital Saint-AntoineParis, France
- *Correspondence: Patricia Forgez, INSERM-UPMC UMR_S938, Hôpital Saint-Antoine, Bâtiment Raoul Kourilsky, 184 rue du Faubourg St-Antoine, 75571 Paris Cedex 12, France. e-mail:
| |
Collapse
|
36
|
Alshoukr F, Prignon A, Brans L, Jallane A, Mendes S, Talbot JN, Tourwé D, Barbet J, Gruaz-Guyon A. Novel DOTA-neurotensin analogues for 111In scintigraphy and 68Ga PET imaging of neurotensin receptor-positive tumors. Bioconjug Chem 2011; 22:1374-85. [PMID: 21662976 DOI: 10.1021/bc200078p] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Overexpression of the high affinity neurotensin receptor 1 (NTSR1), demonstrated in several human cancers, has been proposed as a new marker for human ductal pancreatic carcinoma and as an independent factor for poor prognosis for ductal breast cancer, head and neck squamous cell carcinoma, and non-small cell lung cancer. The aim of the present study was to develop new DOTA-neurotensin analogues for positron emission tomography (PET) imaging with (68)Ga and for targeted radiotherapy with (90)Y or (177)Lu. We synthesized a DOTA-neurotensin analogue series. Two of these peptides bear two sequence modifications for metabolic stability: DOTA-NT-20.3 shares the same peptide sequence as the previously described DTPA-NT-20.3. In the sequence of DOTA-NT-20.4, the Arg(8)-Arg(9) bond was N-methylated instead of the Pro(7)-Arg(8) bond in DOTA-NT-20.3. An additional sequence modification was introduced in DOTA-LB119 to increase stability. A spacer was added between DOTA and the peptide sequence to increase affinity. Binding to HT29 cells, which express NTSR1, in vivo stability, and biodistribution of the various analogues were compared, and the best candidate was used to image tumors of various sizes with the microPET in mice. (111)In-DOTA-NT-20.3, in spite of a relatively high uptake in kidneys, showed specific tumor uptake and elevated tumor to other organ uptake ratios. High contrast images were obtained at early time points after injection that allowed tumor detection at a time interval postinjection appropriate for imaging with the short-lived radionuclide (68)Ga. (111)In-DOTA-NT-20.4 displayed inferior binding to HT29 cells and reduced tumor uptake. (111)In-DOTA-LB119 displayed at early time points a significantly lower renal uptake but also a lower tumor uptake than (111)In-DOTA-NT-20.3, although binding to HT29 cells was similar. (68)Ga-DOTA-NT-20.3 displayed higher tumor uptake than (68)Ga-DOTA-LB119 and allowed the detection of very small tumors by PET. In conclusion, DOTA-NT-20.3 is a promising candidate for (68)Ga-PET imaging of neurotensin receptor-positive tumors. DOTA-NT-20.3 may also be considered for therapy, as the yttrium-labeled peptide has higher affinity than that of the indium-labeled one. A prerequisite for therapeutic application of this neurotensin analogue would be to lower kidney uptake, for example, by infusion of basic amino acids, gelofusin, or albumin fragments, to prevent nephrotoxicity, as with radiolabeled somatostatin analogues.
Collapse
|
37
|
|
38
|
Liu G, Dou S, Baker S, Akalin A, Cheng D, Chen L, Rusckowski M, Hnatowich DJ. A preclinical 188Re tumor therapeutic investigation using MORF/cMORF pretargeting and an antiTAG-72 antibody CC49. Cancer Biol Ther 2011; 10:767-74. [PMID: 21099368 DOI: 10.4161/cbt.10.8.12879] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The utility of MORF/cMORF pretargeting for the radiotherapy of cancer requires further validation in tumored mice before clinical trials. We now report on a therapeutic study in mice pretargeted with MORF-CC49 (an anti-TAG-72 antibody CC49 conjugated with MORF, a phosphorodiamidate morpholino oligomer) and then targeted by 188Re-cMORF (a 188Re labeled complementary MORF). Before the dose-escalating therapeutic study, a pretargeting study in LS174T tumored mice was performed at tracer levels. By both necropsy and imaging, the tracer study showed that the whole body radioactivity was largely restricted to tumor in the mice pretargeted 48 h earlier with MORF-CC49 and the tumor radioactivity was retained over 90 h. After decay correction, a best-fit to the biodistribution provided the areas under the radioactivity curves (AUCs) used for the radiation dose estimates. The tumor to normal organ AUC ratios in all cases were greater than unity and ranged from 3 (kidneys) to 48 (muscle). Tumor growth was inhibited in the therapy study. At the highest 188Re dose of 1.40 mCi, a complete but temporary tumor remission was evident in 3 out of the 5 animals. Histological examination of tissues from these animals showed no evidence of cytotoxicity to normal tissues but obvious radiation damage to tumor. In conclusion, effective radiotherapy was achieved in a mouse model by MORF/cMORF pretargeting using 188Re as the therapeutic radionuclide and CC49 as the pretargeting antibody.
Collapse
Affiliation(s)
- Guozheng Liu
- Division of Nuclear Medicine, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Röhrich A, Bergmann R, Kretzschmann A, Noll S, Steinbach J, Pietzsch J, Stephan H. A novel tetrabranched neurotensin(8-13) cyclam derivative: synthesis, 64Cu-labeling and biological evaluation. J Inorg Biochem 2011; 105:821-32. [PMID: 21497581 DOI: 10.1016/j.jinorgbio.2011.02.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/21/2011] [Accepted: 02/21/2011] [Indexed: 12/11/2022]
Abstract
New macrocyclic 1,4,8,11-tetraazacyclotetradecane (cyclam) derivatives with 1, 2 and 4 neurotensin(8-13) units 4, 5 and 7 have been synthesized. Compounds 4 and 5 were prepared by the reaction of non-stabilized neurotensin(8-13) and cyclamtetrapropionic acid 2 using 1-ethyl-3-(3-dimethylaminocarbonyl)carbodiimide-hydrochloride and N-hydroxysulfosuccinimide. The tetrameric compound 7 was synthesized by Michael addition of neurotensin(8-13) acrylamide 6 and cyclam 1. The copper(II) complexation behavior of 4, 5 and 7 was investigated by UV/visible spectrophotometry and shows that the metal center resides inside the N4 chromophore with additional apical interactions established with pendant arms. The novel tetrabranched NT(8-13) cyclam 7 with nanomolar neurotensin receptor 1 binding affinity was efficiently radiolabeled with (64)Cu under mild conditions. (64)Cu⊂7 showed slow transchelation in the presence of a large amount of cyclam as competing ligand, while it completely remains intact in the presence of EDTA. The in vivo behavior of (64)Cu⊂7 was studied in rats and mice. The metabolic stability in rodent models was high with a half-life of intact (64)Cu⊂7 in plasma of 34 min in rats and 60 min in the mice, respectively. The binding affinity was high enough to demonstrate in vivo binding of (64)Cu⊂7 to NTR1 overexpressing HT-29 tumor xenotransplants in nude mice. Regarding elimination, (64)Cu⊂7 showed a substantial renal and reticuloendothelial accumulation. On the other hand, metabolization of the compound in vivo with a resulting metabolite-postulated to be the (64)Cu-cyclam-tetraarginine complex-also showed long retention in the circulating blood, preventing a better contrast of tumor imaging.
Collapse
Affiliation(s)
- Anika Röhrich
- Institute of Radiopharmacy, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Gromova P, Rubin BP, Thys A, Erneux C, Vanderwinden JM. Neurotensin receptor 1 is expressed in gastrointestinal stromal tumors but not in interstitial cells of Cajal. PLoS One 2011; 6:e14710. [PMID: 21364741 PMCID: PMC3041753 DOI: 10.1371/journal.pone.0014710] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 01/29/2011] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal stromal tumors (GIST) are thought to derive from the interstitial cells of Cajal (ICC) or an ICC precursor. Oncogenic mutations of the KIT or PDGFRA receptor tyrosine kinases are present in the majority of GIST, leading to ligand-independent activation of the intracellular signal transduction pathways. We previously investigated the gene expression profile in the murine KitK641E GIST model and identified Ntsr1 mRNA, encoding the Neurotensin receptor 1, amongst the upregulated genes. Here we characterized Ntsr1 mRNA and protein expression in the murine KitK641E GIST model and in tissue microarrays of human GIST. Ntsr1 mRNA upregulation in KitK641E animals was confirmed by quantitative PCR. Ntsr1 immunoreactivity was not detected in the Kit positive ICC of WT mice, but was present in the Kit positive hyperplasia of KitK641E mice. In the normal human gut, NTSR1 immunoreactivity was detected in myenteric neurons but not in KIT positive ICC. Two independent tissue microarrays, including a total of 97 GIST, revealed NTSR1 immunoreactivity in all specimens, including the KIT negative GIST with PDGFRA mutation. NTSR1 immunoreactivity exhibited nuclear, cytoplasmic or mixed patterns, which might relate to variable levels of NTSR1 activation. As studies using radio-labeled NTSR1 ligand analogues for whole body tumor imaging and for targeted therapeutic interventions have already been reported, this study opens new perspectives for similar approaches in GIST.
Collapse
Affiliation(s)
- Petra Gromova
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Brian P. Rubin
- Anatomic Pathology and Molecular Genetics, Cleveland Clinic, Lerner Research Institute and Taussig Cancer Center, Cleveland, Ohio, United States of America
| | - An Thys
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Christophe Erneux
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jean-Marie Vanderwinden
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- * E-mail:
| |
Collapse
|
41
|
Chen K, Chen X. Design and development of molecular imaging probes. Curr Top Med Chem 2011; 10:1227-36. [PMID: 20388106 DOI: 10.2174/156802610791384225] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 02/27/2010] [Indexed: 01/17/2023]
Abstract
Molecular imaging, the visualization, characterization and measurement of biological processes at the cellular, subcellular level, or even molecular level in living subjects, has rapidly gained importance in the dawning era of personalized medicine. Molecular imaging takes advantage of the traditional diagnostic imaging techniques and introduces molecular imaging probes to determine the expression of indicative molecular markers at different stages of diseases and disorders. As a key component of molecular imaging, molecular imaging probe must be able to specifically reach the target of interest in vivo while retaining long enough to be detected. A desirable molecular imaging probe with clinical translation potential is expected to have unique characteristics. Therefore, design and development of molecular imaging probe is frequently a challenging endeavor for medicinal chemists. This review summarizes the general principles of molecular imaging probe design and some fundamental strategies of molecular imaging probe development with a number of illustrative examples.
Collapse
Affiliation(s)
- Kai Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
42
|
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]
|
43
|
Mindt TL, Struthers H, Spingler B, Brans L, Tourwé D, García-Garayoa E, Schibli R. Molecular Assembly of Multifunctional 99mTc Radiopharmaceuticals Using “Clickable” Amino Acid Derivatives. ChemMedChem 2010; 5:2026-38. [DOI: 10.1002/cmdc.201000342] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
44
|
Maschauer S, Einsiedel J, Hocke C, Hübner H, Kuwert T, Gmeiner P, Prante O. Synthesis of a (68)ga-labeled peptoid-Peptide hybrid for imaging of neurotensin receptor expression in vivo. ACS Med Chem Lett 2010; 1:224-8. [PMID: 24900199 DOI: 10.1021/ml1000728] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 05/14/2010] [Indexed: 12/11/2022] Open
Abstract
The neurotensin receptor subtype 1 (NTS1) represents an attractive molecular target for imaging various tumors. Positron emission tomography (PET) gained widespread importance due to its sensitivity. We combined the design of a metabolically stable neurotensin analogue with a (68)Ga-radiolabeling approach. The (68)Ga-labeled peptoid-peptide hybrid [(68)Ga]3 revealed high stability, specific tumor uptake (0.7%ID/g, 65 min p.i.), and advantageous biokinetics in vivo using HT29 tumor-bearing nude mice. Because of the ability to internalize into NTS1-expressing tumor cells, [(68)Ga]3 proved to be highly suitable for a reliable and practical visualization of NTS1-expressing tumors in vivo by small animal PET.
Collapse
Affiliation(s)
- Simone Maschauer
- Laboratory of Molecular Imaging, Clinic of Nuclear Medicine, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany
| | | | - Carsten Hocke
- Laboratory of Molecular Imaging, Clinic of Nuclear Medicine, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Harald Hübner
- Department of Chemisty and Pharmacy, Emil Fischer Center
| | - Torsten Kuwert
- Laboratory of Molecular Imaging, Clinic of Nuclear Medicine, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemisty and Pharmacy, Emil Fischer Center
| | - Olaf Prante
- Laboratory of Molecular Imaging, Clinic of Nuclear Medicine, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany
| |
Collapse
|
45
|
Synthesis of neurotensin(8–13)–phosphopeptide heterodimers via click chemistry. Bioorg Med Chem Lett 2010; 20:3306-9. [DOI: 10.1016/j.bmcl.2010.04.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 04/07/2010] [Accepted: 04/12/2010] [Indexed: 11/19/2022]
|
46
|
Alshoukr F, Rosant C, Maes V, Abdelhak J, Raguin O, Burg S, Sarda L, Barbet J, Tourwé D, Pelaprat D, Gruaz-Guyon A. Novel neurotensin analogues for radioisotope targeting to neurotensin receptor-positive tumors. Bioconjug Chem 2009; 20:1602-10. [PMID: 19610615 DOI: 10.1021/bc900151z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The increased expression of the neurotensin (NT) receptor NTS1 by different cancer cells, such as pancreatic adenocarcinoma and ductal breast cancer cells, as compared to normal epithelium, offers the opportunity to target these tumors with radiolabeled neurotensin analogues for diagnostic or therapeutic purposes. The aim of the present study was to design and synthesize new neurotensin radioligands and to select a lead molecule with high in vivo tumor selectivity for further development. Two series of neurotensin analogues bearing DTPA were tested: a series of NT(8-13) analogues, with DTPA coupled to the α-NH(2), sharing the same peptide sequence with analogues previously developed for radiolabeling with technetium or rhenium, as well as an NT(6-13) series in which DTPA was coupled to the ε-NH(2) of Lys(6). Changes were introduced to stabilize the bonds between Arg(8)-Arg(9), Pro(10)-Tyr(11), and Tyr(11)-Ile(12) to provide metabolic stability. Structure-activity studies of NT analogues have shown that the attachment of DTPA induces an important loss of affinity unless the distance between the chelator and the NT(8-13) sequence, which binds to the NTS1 receptor, is increased. The doubly stabilized DTPA-NT-20.3 exhibits a high affinity and an elevated stability to enzymatic degradation. It shows specific tumor uptake and high tumor to blood, to liver, and to intestine activity uptake ratios and affords high-contrast planar and SPECT images in an animal model. The DTPA-NT-20.3 peptide is a promising candidate for imaging neurotensin receptor-positive tumors, such as pancreatic adenocarcinoma and invasive ductal breast cancer. Analogues carrying DOTA are being developed for yttrium-90 or lutetium-177 labeling.
Collapse
|
47
|
Myers RM, Shearman JW, Kitching MO, Ramos-Montoya A, Neal DE, Ley SV. Cancer, chemistry, and the cell: molecules that interact with the neurotensin receptors. ACS Chem Biol 2009; 4:503-25. [PMID: 19462983 DOI: 10.1021/cb900038e] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The literature covering neurotensin (NT) and its signalling pathways, receptors, and biological profile is complicated by the fact that the discovery of three NT receptor subtypes has come to light only in recent years. Moreover, a lot of this literature explores NT in the context of the central nervous system and behavioral studies. However, there is now good evidence that the up-regulation of NT is intimately involved in cancer development and progression. This Review aims to summarize the isolation, cloning, localization, and binding properties of the accepted receptor subtypes (NTR1, NTR2, and NTR3) and the molecules known to bind at these receptors. The growing role these targets are playing in cancer research is also discussed. We hope this Review will provide a useful overview and a one-stop resource for new researchers engaged in this field at the chemistry-biology interface.
Collapse
Affiliation(s)
- Rebecca M. Myers
- Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - James W. Shearman
- Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Matthew O. Kitching
- Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Antonio Ramos-Montoya
- CRUK-Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - David E. Neal
- CRUK-Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - Steven V. Ley
- Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|