1
|
Al-Ibraheem A, Allouzi S, Abdlkadir AS, Mikhail-Lette M, Al-Rabi K, Ma'koseh M, Knoll P, Abdelrhman Z, Shahin O, Juweid ME, Paez D, Lopci E. PET/CT in leukemia: utility and future directions. Nucl Med Commun 2024; 45:550-563. [PMID: 38646840 DOI: 10.1097/mnm.0000000000001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
2-Deoxy-2-[ 18 F]fluoro- d -glucose PET/computed tomography ([ 18 F]FDG PET/CT) has proven to be a sensitive method for the detection and evaluation of hematologic malignancies, especially lymphoma. The increasing incidence and mortality rates of leukemia have raised significant concerns. Through the utilization of whole-body imaging, [ 18 F]FDG PET/CT provides a thorough assessment of the entire bone marrow, complementing the limited insights provided by biopsy samples. In this regard, [ 18 F]FDG PET/CT has the ability to assess diverse types of leukemia The utilization of [ 18 F]FDG PET/CT has been found to be effective in evaluating leukemia spread beyond the bone marrow, tracking disease relapse, identifying Richter's transformation, and assessing the inflammatory activity associated with acute graft versus host disease. However, its role in various clinical scenarios in leukemia remains unacknowledged. Despite their less common use, some novel PET/CT radiotracers are being researched for potential use in specific scenarios in leukemia patients. Therefore, the objectives of this review are to provide a thorough assessment of the current applications of [ 18 F]FDG PET/CT in the staging and monitoring of leukemia patients, as well as the potential for an expanding role of PET/CT in leukemia patients.
Collapse
Affiliation(s)
- Akram Al-Ibraheem
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC),
- Department of Radiology and Nuclear Medicine, School of Medicine, University of Jordan, Amman, Jordan,
| | - Sudqi Allouzi
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC),
| | | | - Miriam Mikhail-Lette
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria,
| | - Kamal Al-Rabi
- Department of Medical Oncology, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Mohammad Ma'koseh
- Department of Medical Oncology, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Peter Knoll
- Dosimetry and Medical Radiation Physics Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria,
| | - Zaid Abdelrhman
- Department of Medical Oncology, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Omar Shahin
- Department of Medical Oncology, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Malik E Juweid
- Department of Radiology and Nuclear Medicine, University of Jordan, Amman, Jordan and
| | - Diana Paez
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria,
| | - Egesta Lopci
- Department of Nuclear Medicine, IRCCS - Humanitas Clinical and Research Hospital, Rozzano (MI), Italy
| |
Collapse
|
2
|
van der Wildt B, Nezam M, Kooijman EJ, Reyes ST, Shen B, Windhorst AD, Chin FT. Evaluation of carbon-11 labeled 5-(1-methyl-1H-pyrazol-4-yl)-N-(2-methyl-5-(3-(trifluoromethyl)benzamido)phenyl)nicotinamide as PET tracer for imaging of CSF-1R expression in the brain. Bioorg Med Chem 2021; 42:116245. [DOI: 10.1016/j.bmc.2021.116245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/14/2022]
|
3
|
Bolcaen J, Nair S, Driver CHS, Boshomane TMG, Ebenhan T, Vandevoorde C. Novel Receptor Tyrosine Kinase Pathway Inhibitors for Targeted Radionuclide Therapy of Glioblastoma. Pharmaceuticals (Basel) 2021; 14:626. [PMID: 34209513 PMCID: PMC8308832 DOI: 10.3390/ph14070626] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GB) remains the most fatal brain tumor characterized by a high infiltration rate and treatment resistance. Overexpression and/or mutation of receptor tyrosine kinases is common in GB, which subsequently leads to the activation of many downstream pathways that have a critical impact on tumor progression and therapy resistance. Therefore, receptor tyrosine kinase inhibitors (RTKIs) have been investigated to improve the dismal prognosis of GB in an effort to evolve into a personalized targeted therapy strategy with a better treatment outcome. Numerous RTKIs have been approved in the clinic and several radiopharmaceuticals are part of (pre)clinical trials as a non-invasive method to identify patients who could benefit from RTKI. The latter opens up the scope for theranostic applications. In this review, the present status of RTKIs for the treatment, nuclear imaging and targeted radionuclide therapy of GB is presented. The focus will be on seven tyrosine kinase receptors, based on their central role in GB: EGFR, VEGFR, MET, PDGFR, FGFR, Eph receptor and IGF1R. Finally, by way of analyzing structural and physiological characteristics of the TKIs with promising clinical trial results, four small molecule RTKIs were selected based on their potential to become new therapeutic GB radiopharmaceuticals.
Collapse
Affiliation(s)
- Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Shankari Nair
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Cathryn H. S. Driver
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Brits 0240, South Africa;
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
| | - Tebatso M. G. Boshomane
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Preclinical Drug Development Platform, Department of Science and Technology, North West University, Potchefstroom 2520, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| |
Collapse
|
4
|
Lien VT, Celen S, Nuruddin S, Attili B, Doumont G, Van Simaeys G, Bormans G, Klaveness J, Olberg DE. Preclinical evaluation of [ 18F]cabozantinib as a PET imaging agent in a prostate cancer mouse model. Nucl Med Biol 2021; 93:74-80. [PMID: 33422771 DOI: 10.1016/j.nucmedbio.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Cabozantinib is a tyrosine kinase inhibitor (TKI) approved for the treatment of medullary thyroid cancer, renal cell carcinoma and hepatocellular carcinoma, and is currently in clinical trials for the treatment of prostate cancer and others. It exerts its therapeutic effect mainly through inhibition of the tyrosine kinases MET (hepatocyte growth factor receptor) and VEGFR2 (vascular endothelial growth factor receptor 2), in addition to several other kinases involved in cancer. PET imaging with TKIs such as [18F]cabozantinib could potentially aid in cancer diagnosis and guide treatment. This study aims to evaluate the utility of [18F]cabozantinib as a PET imaging probe in PC3 tumor xenografted mice. METHODS [18F]cabozantinib was evaluated in non-tumor and tumor bearing (PC3 xenografted) male mice by ex vivo biodistribution studies and in vivo μPET imaging. Pretreatment studies were performed in the tumor bearing mice with the MET inhibitor PF04217903. Mouse plasma was analyzed with HPLC to quantify radiometabolites. To further evaluate the binding specificity of [18F]cabozantinib, in vitro autoradiography studies on heart and PC3 tumor sections were performed in the presence of authentic cabozantinib or specific MET and VEGFR2 inhibitors. RESULTS Tissue distribution studies in non-tumor bearing mice revealed slow blood clearance, absence of brain uptake and a high myocardial uptake. In the tumor bearing mice, tumor uptake was low (0.58 ± 0.20% ID/g at 30 min post tracer injection), which was confirmed by μPET imaging. No differences in tissue distribution and kinetics were observed in both biodistributions and μPET studies after pretreatment with the MET inhibitor PF04217903. At 30 min post tracer injection, 60 ± 3% of the recovered radioactivity in plasma in non-tumor bearing mice was present as intact tracer. [18F]cabozantinib binding in vitro to heart and tumor tissues was partly blocked in the presence of selective MET and VEGFR2 inhibitors (up to 40% block). The fraction of non-specific binding was relatively high for both tissues (66% for heart and 39% for tumor). CONCLUSION [18F]cabozantinib exhibits non-favorable properties as a PET imaging probe, demonstrated by slow excretion kinetics along with low tumor uptake and high non-specific binding in tumor and heart tissue. The results reflect cabozantinibs multi-kinase activity, making PET imaging of tumor specific kinase expression with [18F]cabozantinib challenging.
Collapse
Affiliation(s)
- Vegard Torp Lien
- Department of Pharmacy, University of Oslo, Boks 1068, Blindern, 0316 Oslo, Norway; Norwegian Medical Cyclotron Center, Oslo, Norway.
| | - Sofie Celen
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | | | - Bala Attili
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Gilles Doumont
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), Charleroi, Belgium
| | - Gaetan Van Simaeys
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), Charleroi, Belgium; Department of Nuclear Medicine, Erasme University Hospital, Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Jo Klaveness
- Department of Pharmacy, University of Oslo, Boks 1068, Blindern, 0316 Oslo, Norway
| | - Dag Erlend Olberg
- Department of Pharmacy, University of Oslo, Boks 1068, Blindern, 0316 Oslo, Norway; Norwegian Medical Cyclotron Center, Oslo, Norway
| |
Collapse
|
5
|
Cai H, Shi Q, Tang Y, Chen L, Chen Y, Tao Z, Yang H, Xie F, Wu X, Liu N, Yang Y, Wu H, Tian R, Lu X, Li L. Positron Emission Tomography Imaging of Platelet-Derived Growth Factor Receptor β in Colorectal Tumor Xenograft Using Zirconium-89 Labeled Dimeric Affibody Molecule. Mol Pharm 2019; 16:1950-1957. [PMID: 30986347 DOI: 10.1021/acs.molpharmaceut.8b01317] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Huawei Cai
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiuxiao Shi
- Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Tang
- Key Lab of Radiation Physics and Technology, Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| | - Lihong Chen
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yue Chen
- Departments of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ze Tao
- Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Yang
- Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaoai Wu
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Nan Liu
- Departments of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yuanyou Yang
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Haoxing Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital and West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Rong Tian
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaofeng Lu
- Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Li
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Effendi N, Mishiro K, Takarada T, Makino A, Yamada D, Kitamura Y, Shiba K, Kiyono Y, Odani A, Ogawa K. Radiobrominated benzimidazole-quinoline derivatives as Platelet-derived growth factor receptor beta (PDGFRβ) imaging probes. Sci Rep 2018; 8:10369. [PMID: 29991770 PMCID: PMC6039436 DOI: 10.1038/s41598-018-28529-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/25/2018] [Indexed: 12/31/2022] Open
Abstract
Platelet-derived growth factor receptor beta (PDGFRβ) affects in numerous human cancers and has been recognized as a promising molecular target for cancer therapies. The overexpression of PDGFRβ could be a biomarker for cancer diagnosis. Radiolabeled ligands having high affinity for the molecular target could be useful tools for the imaging of overexpressed receptors in tumors. In this study, we aimed to develop radiobrominated PDGFRβ ligands and evaluate their effectiveness as PDGFRβ imaging probes. The radiolabeled ligands were designed by modification of 1-{2-[5-(2-methoxyethoxy)-1H- benzo[d]imidazol-1-yl]quinolin-8-yl}piperidin-4-amine (1), which shows selective inhibition profile toward PDGFRβ. The bromine atom was introduced directly into C-5 of the quinoline group of 1, or indirectly by the conjugation of 1 with the 3-bromo benzoyl group. [77Br]1-{5-Bromo-2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]quinoline-8-yl}piperidin-4-amine ([77Br]2) and [77Br]-N-3-bromobenzoyl-1-{2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]quinolin-8-yl}-piperidin-4-amine ([77Br]3) were prepared using a bromodestannylation reaction. In a cellular uptake study, [77Br]2 and [77Br]3 more highly accumulatd in BxPC3-luc cells (PDGFRβ-positive) than in MCF7 cells (PDGFRβ-negative), and their accumulation was significantly reduced by pretreatment with inhibitors. In biodistribution experiments, [77Br]2 accumulation was higher than [77Br]3 accumulation at 1 h postinjection. These findings suggest that [76Br]2 is more promising for positron emission tomography (PET) imaging of PDGFRβ than [76Br]3.
Collapse
Affiliation(s)
- Nurmaya Effendi
- Kanazawa University, Graduate School of Pharmaceutical Sciences, Kakuma-machi, Kanazawa, 920-1192, Japan
- Universitas Muslim Indonesia, Faculty of Pharmacy, Urip Sumiharjo KM. 10, Makassar, 90-231, Indonesia
| | - Kenji Mishiro
- Kanazawa University, Institute for Frontier Science Initiative, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Takeshi Takarada
- Okayama University, Graduate School of Medicine, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Akira Makino
- University of Fukui, Biomedical Imaging Research Center, 23-3 Matsuoka Shimoaizuki, Yoshida, 910-1193, Japan
| | - Daisuke Yamada
- Okayama University, Graduate School of Medicine, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Yoji Kitamura
- Kanazawa University, Advanced Science Research Centre, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Kazuhiro Shiba
- Kanazawa University, Advanced Science Research Centre, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Yasushi Kiyono
- University of Fukui, Biomedical Imaging Research Center, 23-3 Matsuoka Shimoaizuki, Yoshida, 910-1193, Japan
| | - Akira Odani
- Kanazawa University, Graduate School of Pharmaceutical Sciences, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Kazuma Ogawa
- Kanazawa University, Graduate School of Pharmaceutical Sciences, Kakuma-machi, Kanazawa, 920-1192, Japan.
- Kanazawa University, Institute for Frontier Science Initiative, Kakuma-machi, Kanazawa, 920-1192, Japan.
| |
Collapse
|
7
|
Abstract
The process of discovering and developing a new pharmaceutical is a long, difficult, and risky process that requires numerous resources. Molecular imaging techniques such as PET have recently become a useful tool for making decisions along a drug candidate's development timeline. PET is a translational, noninvasive imaging technique that provides quantitative information about a potential drug candidate and its target at the molecular level. Using this technique provides decisional information to ensure that the right drug candidate is being chosen, for the right target, at the right dose within the right patient population. This review will focus on small molecule PET tracers and how they are used within the drug discovery process. PET provides key information about a drug candidate's pharmacokinetic and pharmacodynamic properties in both preclinical and clinical studies. PET is being used in all phases of the drug discovery and development process, and the goal of these studies are to accelerate the process in which drugs are developed.
Collapse
Affiliation(s)
- David J Donnelly
- Bristol-Myers Squibb Pharmaceutical Research and Development, Princeton, NJ.
| |
Collapse
|
8
|
Wagner M, Wuest M, Hamann I, Lopez-Campistrous A, McMullen TPW, Wuest F. Molecular imaging of platelet-derived growth factor receptor-alpha (PDGFRα) in papillary thyroid cancer using immuno-PET. Nucl Med Biol 2017; 58:51-58. [PMID: 29367096 DOI: 10.1016/j.nucmedbio.2017.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Receptor tyrosine kinase (RTK) platelet-derived growth factor receptor-alpha (PDGFRα) was recently identified as a molecular switch for dedifferentiation in thyroid cancer that predicts resistance to therapy as well as recurrence of disease in papillary thyroid cancer. Here we describe the radiolabeling and functional characterization of an imaging probe based on a PDGFRα-specific monoclonal antibody (mAb) for immuno-PET imaging of PDGFRα in papillary thyroid cancer. METHODS Antibody D13C6 (Cell Signaling) was decorated with chelator NOTA using bioconjugation reaction with 2-(p-NCS-Bz)-NOTA. Radiolabeling was carried out using 40 μg of antibody-NOTA conjugate with 143-223 MBq of [64Cu]CuCl2 in 0.25 M NaOAc (pH 5.5) at 30 °C for 1 h. The reaction mixture was purified with size-exclusion chromatography (PD-10 column). PDGFRα and mock transfected B-CPAP thyroid cancer cells lines for validation of 64Cu-labeled immuno-conjugates were generated using LVX-Tet-On technology. PET imaging was performed in NSG mice bearing bilaterally-induced PDGFRα (+/-) B-CPAP tumors. RESULTS Bioconjugation of NOTA chelator to monoclonal antibody D13C6 resulted in 2.8 ± 1.3 chelator molecules per antibody as determined by radiometric titration with 64Cu. [64Cu]Cu-NOTA-D13C6 was isolated in high radiochemical purity (>98%) and good radiochemical yields (19-61%). The specific activity was 0.9-5.1 MBq/μg. Cellular uptake studies revealed a specific radiotracer uptake in PDGFRα expressing cells compared to control cells. PET imaging resulted in SUVmean values of ~5.5 for PDGFRα (+) and ~2 for PDGFRα (-) tumors, after 48 h p.i.. After 1 h, radiotracer uptake was also observed in the bone marrow (SUVmean ~5) and spleen (SUVmean ~8.5). CONCLUSION Radiolabeled antibody [64Cu]Cu-NOTA-D13C6 represents a novel and promising radiotracer for immuno-PET imaging of PDGFRα in metastatic papillary thyroid cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE The presented work has the potential to allow physicians to identify papillary thyroid cancer patients at risk of metastases by using the novel immuno-PET imaging assay based on PDGFRα-targeting antibody [64Cu]Cu-NOTA-D13C6.
Collapse
Affiliation(s)
- Michael Wagner
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada
| | - Melinda Wuest
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada
| | - Ingrit Hamann
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada
| | - Ana Lopez-Campistrous
- University of Alberta, Department of Surgery, 2D4.41 Walter Mackenzie Centre 8440- 112 Street, Edmonton, AB T6G 2B7, Canada
| | - Todd P W McMullen
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada; University of Alberta, Department of Surgery, 2D4.41 Walter Mackenzie Centre 8440- 112 Street, Edmonton, AB T6G 2B7, Canada.
| | - Frank Wuest
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada.
| |
Collapse
|
9
|
Synthesis, in vitro and in vivo evaluation of 18F-fluoronorimatinib as radiotracer for Imatinib-sensitive gastrointestinal stromal tumors. Nucl Med Biol 2017; 57:1-11. [PMID: 29175467 DOI: 10.1016/j.nucmedbio.2017.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/03/2017] [Accepted: 11/15/2017] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Gastrointestinal stromal tumors (GIST) have a wide range of mutations, but can mostly be treated with Imatinib, until eventually resistance towards this tyrosine kinase inhibitor is acquired. Early and non-invasive determination of the sensitivity of the tumor and its metastases towards Imatinib by positron emission tomography (PET) would be beneficial for therapy planning and monitoring. METHODS We developed a synthesis strategy towards the precursor molecule, performed the 18F-synthesis and in the following evaluated the radioligand in vitro regarding its lipophilicity, stability and biological activity (KIT binding properties) as well as its in vivo properties in GIST tumor-bearing mice. RESULTS [18F]fluoronorimatinib could be obtained in an overall radiochemical yield of 22.2±3.3% within 90min. The radioligand showed high GIST cell uptake and was able to distinguish between Imatinib-sensitive and resistant tumor cell lines (GIST-T1, GIST882, GIST430) in vitro. Further biological evaluations of the ligand towards 9 different GIST-relevant KIT mutations showed comparable binding affinities compared to the structural lead Norimatinib (65nM vs. 53nM for wt-KIT). The in vivo evaluation of the newly developed radioligand showed tumor-to-background-ratios comparable to previously described, similar radiotracers. CONCLUSIONS Thus, [18F]fluoronorimatinib is able to distinguish between Imatinib-resistant and sensitive KIT mutations. Although no improvement of in vivo tumor-to-background ratios could be achieved compared to formerly described radioligands, the hepatic uptake could be considerably reduced, being advantageous for the imaging of GIST. Advances in knowledge and implications for patient care: We were able to show that it is possible to significantly reduce the unfavorably high hepatic uptake of small-molecule radioligands applicable for GIST PET imaging. This work can thus be the basis for further work intending to develop a PET-radioligand for Imatinib-dependent GIST imaging.
Collapse
|
10
|
Hrynchak I, Sousa E, Pinto M, Costa VM. The importance of drug metabolites synthesis: the case-study of cardiotoxic anticancer drugs. Drug Metab Rev 2017; 49:158-196. [DOI: 10.1080/03602532.2017.1316285] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ivanna Hrynchak
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Emília Sousa
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- CIIMAR – Centro Interdisciplinar de Investigação Marinha e Ambiental, Matosinhos, Portugal
| | - Madalena Pinto
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- CIIMAR – Centro Interdisciplinar de Investigação Marinha e Ambiental, Matosinhos, Portugal
| | - Vera Marisa Costa
- Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, UCIBIO, REQUIMTE (Rede de Química e Tecnologia), Universidade do Porto, Porto, Portugal
| |
Collapse
|
11
|
Stolarczyk EU, Eksanow K, Filip K. Determination of Three Potential Genotoxic Impurities in Imatinib Mesylate by Gas Chromatography—Mass Spectrometry. ANAL LETT 2016. [DOI: 10.1080/00032719.2016.1146737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
12
|
Recent Advances in the Development and Application of Radiolabeled Kinase Inhibitors for PET Imaging. Molecules 2015; 20:22000-27. [PMID: 26690113 PMCID: PMC6332294 DOI: 10.3390/molecules201219816] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/18/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022] Open
Abstract
Over the last 20 years, intensive investigation and multiple clinical successes targeting protein kinases, mostly for cancer treatment, have identified small molecule kinase inhibitors as a prominent therapeutic class. In the course of those investigations, radiolabeled kinase inhibitors for positron emission tomography (PET) imaging have been synthesized and evaluated as diagnostic imaging probes for cancer characterization. Given that inhibitor coverage of the kinome is continuously expanding, in vivo PET imaging will likely find increasing applications for therapy monitoring and receptor density studies both in- and outside of oncological conditions. Early investigated radiolabeled inhibitors, which are mostly based on clinically approved tyrosine kinase inhibitor (TKI) isotopologues, have now entered clinical trials. Novel radioligands for cancer and PET neuroimaging originating from novel but relevant target kinases are currently being explored in preclinical studies. This article reviews the literature involving radiotracer design, radiochemistry approaches, biological tracer evaluation and nuclear imaging results of radiolabeled kinase inhibitors for PET reported between 2010 and mid-2015. Aspects regarding the usefulness of pursuing selective vs. promiscuous inhibitor scaffolds and the inherent challenges associated with intracellular enzyme imaging will be discussed.
Collapse
|
13
|
Nicolaou KC, Vourloumis D, Totokotsopoulos S, Papakyriakou A, Karsunky H, Fernando H, Gavrilyuk J, Webb D, Stepan AF. Synthesis and Biopharmaceutical Evaluation of Imatinib Analogues Featuring Unusual Structural Motifs. ChemMedChem 2015; 11:31-7. [PMID: 26585829 DOI: 10.1002/cmdc.201500510] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Indexed: 11/10/2022]
Abstract
A convenient synthesis of imatinib, a potent inhibitor of ABL1 kinase and widely prescribed drug for the treatment of a variety of leukemias, was devised and applied to the construction of a series of novel imatinib analogues featuring a number of non-aromatic structural motifs in place of the parent molecule's phenyl moiety. These analogues were subsequently evaluated for their biopharmaceutical properties (e.g., ABL1 kinase inhibitory activity, cytotoxicity). The bicyclo[1.1.1]pentane- and cubane-containing analogues were found to possess higher themodynamic solubility, whereas cubane- and cyclohexyl-containing analogues exhibited the highest inhibitory activity against ABL1 kinase and the most potent cytotoxicity values against cancer cell lines K562 and SUP-B15. Molecular modeling was employed to rationalize the weak activity of the compounds against ABL1 kinase, and it is likely that the observed cytotoxicity of these agents arises through off-target effects.
Collapse
Affiliation(s)
- Kyriacos C Nicolaou
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX, 77005, USA.
| | - Dionisios Vourloumis
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX, 77005, USA. , .,Laboratory of Chemical Biology of Natural Products & Designed Molecules, National Centre of Scientific Research (NCSR) "Demokritos", Agia Paraskevi, Athens, 15310, Greece. ,
| | | | - Athanasios Papakyriakou
- Laboratory of Chemical Biology of Natural Products & Designed Molecules, National Centre of Scientific Research (NCSR) "Demokritos", Agia Paraskevi, Athens, 15310, Greece
| | - Holger Karsunky
- Stemcentrx, Inc., 450 East Jamie Court, San Francisco, CA, 94080, USA
| | - Hanan Fernando
- Stemcentrx, Inc., 450 East Jamie Court, San Francisco, CA, 94080, USA
| | - Julia Gavrilyuk
- Stemcentrx, Inc., 450 East Jamie Court, San Francisco, CA, 94080, USA
| | - Damien Webb
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, MA, 02139, USA
| | - Antonia F Stepan
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
14
|
Abstract
Targeted therapy is gaining prominence in the management of different cancers. Given different mechanism of action compared with traditional chemoradiotherapy, selection of patients for targeted therapy and monitoring response to these agents is difficult with conventional imaging. Various new PET radiopharmaceuticals have been evaluated for molecular imaging of these targets to achieve specific patient selection and response monitoring. These PET/computed tomography (CT) agents target the cell surface receptors, hormone receptors, receptor tyrosine kinases, or angiogenesis components. This article reviews the established and potential role of PET/CT with new radiopharmaceuticals for guiding targeted therapy.
Collapse
Affiliation(s)
- Punit Sharma
- Department of Nuclear Medicine and PET/CT, Eastern Diagnostics India Ltd, 13C Mirza Ghalib Street, Kolkata 700016, India; Diagnostic Nuclear Medicine Division, Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Rakesh Kumar
- Diagnostic Nuclear Medicine Division, Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| | - Abass Alavi
- Division of Nuclear Medicine, Department of Radiology, University of Pennsylvania School of Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Mammatas LH, Verheul HMW, Hendrikse NH, Yaqub M, Lammertsma AA, Menke-van der Houven van Oordt CW. Molecular imaging of targeted therapies with positron emission tomography: the visualization of personalized cancer care. Cell Oncol (Dordr) 2014; 38:49-64. [PMID: 25248503 DOI: 10.1007/s13402-014-0194-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Molecular imaging has been defined as the visualization, characterization and measurement of biological processes at the molecular and cellular level in humans and other living systems. In oncology it enables to visualize (part of) the functional behaviour of tumour cells, in contrast to anatomical imaging that focuses on the size and location of malignant lesions. Available molecular imaging techniques include single photon emission computed tomography (SPECT), positron emission tomography (PET) and optical imaging. In PET, a radiotracer consisting of a positron emitting radionuclide attached to the biologically active molecule of interest is administrated to the patient. Several approaches have been undertaken to use PET for the improvement of personalized cancer care. For example, a variety of radiolabelled ligands have been investigated for intratumoural target identification and radiolabelled drugs have been developed for direct visualization of the biodistibution in vivo, including intratumoural therapy uptake. First indications of the clinical value of PET for target identification and response prediction in oncology have been reported. This new imaging approach is rapidly developing, but uniformity of scanning processes, standardized methods for outcome evaluation and implementation in daily clinical practice are still in progress. In this review we discuss the available literature on molecular imaging with PET for personalized targeted treatment strategies. CONCLUSION Molecular imaging with radiolabelled targeted anticancer drugs has great potential for the improvement of personalized cancer care. The non-invasive quantification of drug accumulation in tumours and normal tissues provides understanding of the biodistribution in relation to therapeutic and toxic effects.
Collapse
Affiliation(s)
- Lemonitsa H Mammatas
- Dept of Medical Oncology VUmc Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
16
|
Strand J, Varasteh Z, Eriksson O, Abrahmsen L, Orlova A, Tolmachev V. Gallium-68-labeled affibody molecule for PET imaging of PDGFRβ expression in vivo. Mol Pharm 2014; 11:3957-64. [PMID: 24972112 DOI: 10.1021/mp500284t] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Platelet-derived growth factor receptor β (PDGFRβ) is a transmembrane tyrosine kinase receptor involved, for example, in angiogenesis. Overexpression and excessive signaling of PDGFRβ has been observed in multiple malignant tumors and fibrotic diseases, making this receptor a pharmaceutical target for monoclonal antibodies and tyrosine kinase inhibitors. Successful targeted therapy requires identification of responding patients. Radionuclide molecular imaging would enable determination of the PDGFRβ status in all lesions using a single noninvasive repeatable procedure. Recently, we have demonstrated that the affibody molecule Z09591 labeled with (111)In can specifically target PDGFRβ-expressing tumors in vivo. The use of positron emission tomography (PET) as an imaging technique would provide superior resolution, sensitivity, and quantitation accuracy. In this study, a DOTA-conjugated Z09591 was labeled with the generator-produced positron emitting radionuclide (68)Ga (T1/2 = 67.6 min, Eβ + max = 1899 keV, 89% β(+)). (68)Ga-DOTA-Z09591 retained the capacity to specifically bind to PDGFRβ-expressing U-87 MG glioma cells. The half-maximum inhibition concentration (IC50) of (68)Ga-DOTA-Z09591 (6.6 ± 1.4 nM) was somewhat higher than that of (111)In-DOTA-Z09591 (1.4 ± 1.2 nM). (68)Ga-DOTA-Z09591 demonstrated specific (saturable) targeting of U-87 MG xenografts in immunodeficient mice. The tumor uptake at 2 h after injection was 3.7 ± 1.7% IA/g, which provided a tumor-to-blood ratio of 8.0 ± 3.1. The only organ with higher accumulation of radioactivity was the kidney. MicroPET imaging provided high-contrast imaging of U-87 MG xenografts. In conclusion, the (68)Ga-labeled affibody molecule Z09591 is a promising candidate for further development as a probe for imaging PDGFRβ expression in vivo using PET.
Collapse
Affiliation(s)
- Joanna Strand
- Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University , Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
17
|
Juszczak K, Maciukiewicz P, Drewa T, Thor PJ. Cajal-like interstitial cells as a novel target in detrusor overactivity treatment: true or myth? Cent European J Urol 2014; 66:413-7. [PMID: 24757530 PMCID: PMC3992455 DOI: 10.5173/ceju.2013.04.art5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/18/2013] [Accepted: 11/25/2013] [Indexed: 11/22/2022] Open
Abstract
Introduction The Cajal–like intestitial cells (ICCs) act as a pacemaker and are responsible for generating smooth muscle activity in the gastrointestinal tract (GI). Interstitial cells that resemble ICCs in the GI have been identified in the urinary bladder. Materials and methods This review is based on a systemic literature research. The medline/pubmed, scopus, embase, and Web of Science databases were browsed in order to identify original and review articles, as well as editorials relating to cajal–like cells, urinary bladder, detrusor overactivity, overactive bladder, glivec, etc. The controlled vocabulary of the Medical Subject Headings (MeSH) database was used to ensure the sensitivity of the searches. 40 papers met the criteria and were used for this review. Results Cajal cells lie in close proximity to the muscle cells, autonomic nerve endings, and urothelial cells. There is increasing evidence that ICCs play role in urinary tract dysfunction development (e.g. detrusor overactivity, primary obstructive megaureter, congenital ureteropelvic junction obstruction, etc.). ICCs may be responsible for generating electrical potentials and induction of detrusor muscle contractions. Novel pathomechanisms of detrusor overactivity development have been postulated, as follows: 1) the disturbance of spontaneous contractility caused by altered signal transduction of ICCs between nerves and detrusor muscle cells, and 2). the alteration in signal transduction between urothelium and afferent nerve endings via suburothelial ICCs. The c–kit receptor is not only a detection marker of these cells, but may also play a crucial role in the control of bladder function. Conclusions Cajal cells in urinary bladder suggest that the c–kit receptor may provide a novel target for treating detrusor overactivity. The review presents the current knowledge of ICCs, its role in urinary bladder function, and potential novel therapeutic strategy.
Collapse
Affiliation(s)
- Kajetan Juszczak
- Department of Pathophysiology, Jagiellonian University, Medical College, Cracow, Poland ; Department of Urology, Memorial Rydygier Hospital, Cracow, Poland
| | | | - Tomasz Drewa
- Department of Tissue Engineering, Medical College, Nicolaus Copernicus University, Toruń, Poland ; Department of Urology, Nicolaus Copernicus University, Toruń, Poland
| | - Piotr J Thor
- Department of Pathophysiology, Jagiellonian University, Medical College, Cracow, Poland
| |
Collapse
|
18
|
Tolmachev V, Varasteh Z, Honarvar H, Hosseinimehr SJ, Eriksson O, Jonasson P, Frejd FY, Abrahmsen L, Orlova A. Imaging of platelet-derived growth factor receptor β expression in glioblastoma xenografts using affibody molecule 111In-DOTA-Z09591. J Nucl Med 2014; 55:294-300. [PMID: 24408895 DOI: 10.2967/jnumed.113.121814] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
UNLABELLED The overexpression and excessive signaling of platelet-derived growth factor receptor β (PDGFRβ) has been detected in cancers, atherosclerosis, and a variety of fibrotic diseases. Radionuclide in vivo visualization of PDGFRβ expression might help to select PDGFRβ targeting treatment for these diseases. The goal of this study was to evaluate the feasibility of in vivo radionuclide imaging of PDGFRβ expression using an Affibody molecule, a small nonimmunoglobulin affinity protein. METHODS The PDGFRβ-binding Z09591 Affibody molecule was site-specifically conjugated with a maleimido derivative of DOTA and labeled with (111)In. Targeting of the PDGFRβ-expressing U-87 MG glioblastoma cell line using (111)In-DOTA-Z09591 was evaluated in vitro and in vivo. RESULTS DOTA-Z09591 was stably labeled with (111)In with preserved specific binding to PDGFRβ-expressing cells in vitro. The dissociation constant for (111)In-DOTA-Z09591 binding to U-87 MG cells was determined to be 92 ± 10 pM. In mice bearing U-87 MG xenografts, the tumor uptake of (111)In-DOTA-Z09591 was 7.2 ± 2.4 percentage injected dose per gram and the tumor-to-blood ratio was 28 ± 14 at 2 h after injection. In vivo receptor saturation experiments demonstrated that targeting of U-87 MG xenografts in mice was PDGFRβ-specific. U-87 MG xenografts were clearly visualized using small-animal SPECT/CT at 3 h after injection. CONCLUSION This study demonstrates the feasibility of in vivo visualization of PDGFRβ-expressing xenografts using an Affibody molecule. Further development of radiolabeled Affibody molecules might provide a useful clinical imaging tool for PDGFRβ expression during various pathologic conditions.
Collapse
Affiliation(s)
- Vladimir Tolmachev
- Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
van der Veldt AAM, Smit EF, Lammertsma AA. Positron Emission Tomography as a Method for Measuring Drug Delivery to Tumors in vivo: The Example of [(11)C]docetaxel. Front Oncol 2013; 3:208. [PMID: 23986880 PMCID: PMC3742054 DOI: 10.3389/fonc.2013.00208] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/30/2013] [Indexed: 01/26/2023] Open
Abstract
Systemic anticancer treatments fail in a substantial number of patients. This may be caused by inadequate uptake and penetration of drugs in malignant tumors. Consequently, improvement of drug delivery to solid tumors may enhance its efficacy. Before evaluating strategies to enhance drug uptake in tumors, better understanding of drug delivery to human tumors is needed. Positron emission tomography (PET) is an imaging technique that can be used to monitor drug pharmacokinetics non-invasively in patients, based on radiolabeling these drugs with short-lived positron emitters. In this mini review, principles and potential applications of PET using radiolabeled anticancer drugs will be discussed with respect to personalized treatment planning in oncology. In particular, it will be discussed how these radiolabeled anticancer drugs could help to develop strategies for improved drug delivery to solid tumors. The development and clinical implementation of PET using radiolabeled anticancer drugs will be illustrated by validation studies of carbon-11 labeled docetaxel ([(11)C]docetaxel) in lung cancer patients.
Collapse
Affiliation(s)
- Astrid A M van der Veldt
- Department of Internal Medicine, VU University Medical Center , Amsterdam , Netherlands ; Department of Radiology and Nuclear Medicine, VU University Medical Center , Amsterdam , Netherlands
| | | | | |
Collapse
|
20
|
Poot AJ, van der Wildt B, Stigter-van Walsum M, Rongen M, Schuit RC, Hendrikse NH, Eriksson J, van Dongen GAMS, Windhorst AD. [¹¹C]Sorafenib: radiosynthesis and preclinical evaluation in tumor-bearing mice of a new TKI-PET tracer. Nucl Med Biol 2013; 40:488-97. [PMID: 23522977 DOI: 10.1016/j.nucmedbio.2013.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/07/2013] [Accepted: 02/02/2013] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Tyrosine kinase inhibitors (TKIs) like sorafenib are important anticancer therapeutics with thus far limited treatment response rates in cancer patients. Positron emission tomography (PET) could provide the means for selection of patients who might benefit from TKI treatment, if suitable PET tracers would be available. The aim of this study was to radiolabel sorafenib (1) with carbon-11 and to evaluate its potential as TKI-PET tracer in vivo. METHODS Synthetic methods were developed in which sorafenib was labeled at two different positions, followed by a metabolite analysis in rats and a PET imaging study in tumor-bearing mice. RESULTS [methyl-(11)C]-1 and [urea-(11)C]-1 were synthesized in yields of 59% and 53%, respectively, with a purity of >99%. The identity of the products was confirmed by coinjection on HPLC with reference sorafenib. In an in vivo metabolite analysis [(11)C]sorafenib proved to be stable. The percentage of intact product in blood-plasma after 45 min was 90% for [methyl-(11)C]-1 and 96% for [urea-(11)C]-1, respectively. Due to the more reliable synthesis, further research regarding PET imaging was performed with [methyl-(11)C]-1 in nude mice bearing FaDu (head and neck cancer), MDA-MB-231 (breast cancer) or RXF393 (renal cancer) xenografts. Highest tracer accumulation at a level of 2.52 ± 0.33%ID/g was observed in RXF393, a xenograft line extensively expressing the sorafenib target antigen Raf-1 as assessed by immunohistochemistry. CONCLUSION In conclusion, we have synthesized [(11)C]sorafenib as PET tracer, which is stable in vivo and has the capability to be used as PET tracer for imaging in tumor-bearing mice.
Collapse
Affiliation(s)
- Alex J Poot
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Deadman BJ, Hopkin MD, Baxendale IR, Ley SV. The synthesis of Bcr-Abl inhibiting anticancer pharmaceutical agents imatinib, nilotinib and dasatinib. Org Biomol Chem 2013; 11:1766-800. [DOI: 10.1039/c2ob27003j] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
22
|
Li J, Huang Y, Huang L, Ye L, Zhou Z, Xiang G, Xu L. Determination of imatinib mesylate and related compounds by field amplified sample stacking with large volume sample injection capillary electrophoresis. J Pharm Biomed Anal 2012; 70:26-31. [DOI: 10.1016/j.jpba.2012.05.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 05/06/2012] [Accepted: 05/07/2012] [Indexed: 11/30/2022]
|
23
|
Ye L, Huang Y, Li J, Xiang G, Xu L. Nonaqueous capillary electrophoresis of imatinib mesylate and related substances. J Sep Sci 2012; 35:2108-13. [DOI: 10.1002/jssc.201200114] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/22/2012] [Accepted: 04/23/2012] [Indexed: 12/19/2022]
Affiliation(s)
| | | | | | - Guangya Xiang
- Tongji School of Pharmacy; Huazhong University of Science and Technology; Wuhan China
| | - Li Xu
- Tongji School of Pharmacy; Huazhong University of Science and Technology; Wuhan China
| |
Collapse
|
24
|
Slobbe P, Poot AJ, Windhorst AD, van Dongen GAMS. PET imaging with small-molecule tyrosine kinase inhibitors: TKI-PET. Drug Discov Today 2012; 17:1175-87. [PMID: 22766374 DOI: 10.1016/j.drudis.2012.06.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/18/2012] [Accepted: 06/21/2012] [Indexed: 12/20/2022]
Abstract
The discovery and increased understanding of tumor targets has led to the development and approval of 12 small molecule tyrosine kinase inhibitors (TKIs). Despite tremendous efforts in TKI development, treatment efficacies with these therapeutics are still too low and improvements require a personalized medicine approach. Positron emission tomography (PET) with radiolabeled TKIs (TKI-PET) is a tracking, quantification and imaging method, which provides a unique understanding of the behavior of these drugs in vivo and of the interaction with their target(s). In this article we provide an overview of tracer synthesis and development because each TKI requires a tailor made approach. Moreover, we describe current preclinical work and the first proof-of-principle clinical studies on the application of TKI-PET, illustrating the potential of this approach for improving therapy efficacy and personalized cancer treatment.
Collapse
Affiliation(s)
- Paul Slobbe
- Department of Nuclear Medicine and PET Research, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
25
|
Shukla S, Chen ZS, Ambudkar SV. Tyrosine kinase inhibitors as modulators of ABC transporter-mediated drug resistance. Drug Resist Updat 2012; 15:70-80. [PMID: 22325423 DOI: 10.1016/j.drup.2012.01.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 01/12/2012] [Accepted: 01/13/2012] [Indexed: 12/23/2022]
Abstract
Tyrosine kinases (TKs) are involved in key signaling events/pathways that regulate cancer cell proliferation, apoptosis, angiogenesis and metastasis. Deregulated activity of TKs has been implicated in several types of cancers. In recent years, tyrosine kinase inhibitors (TKIs) have been developed to inhibit specific kinases whose constitutive activity results in specific cancer types. These TKIs have been found to demonstrate effective anticancer activity and some of them have been approved by the Food and Drug Administration for clinical use or are in clinical trials. However, these targeted therapeutic agents are also transported by ATP-binding cassette (ABC) transporters, resulting in altered pharmacokinetics or development of resistance to these drugs in cancer patients. This review covers the recent findings on the interactions of clinically important TKIs with ABC drug transporters. Future research efforts in the development of novel TKIs with specific targets, seeking improved activity, should consider these underlying causes of resistance to TKIs in cancer cells.
Collapse
Affiliation(s)
- Suneet Shukla
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
26
|
PET imaging with radiolabeled antibodies and tyrosine kinase inhibitors: immuno-PET and TKI-PET. Tumour Biol 2012; 33:607-15. [PMID: 22270450 PMCID: PMC3342498 DOI: 10.1007/s13277-012-0316-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 01/03/2012] [Indexed: 10/26/2022] Open
Abstract
During the last decade, the discovery of critical tumor targets has boosted the design of targeted therapeutic agents with monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs) receiving most of the attention. Immuno-positron emission tomography (immuno-PET) and TKI-PET, the in vivo tracking and quantification of mAbs and TKIs biodistribution with PET, are exciting novel options for better understanding of the in vivo behavior and efficacy of these targeted drugs in individual patients and for more efficient drug development. Very recently, current good manufacturing practice compliant procedures for labeling of mAbs with positron emitters have been described, as well as the preparation of some radiolabeled TKIs, while the first proof of principle studies has been performed in patients. In this review, technical developments in immuno-PET and TKI-PET are described, and their clinical potential is discussed. An overview is provided for the most appealing preclinical immuno-PET and TKI-PET studies, as well as the first clinical achievements with these emerging technologies.
Collapse
|
27
|
Kamal A, Dastagiri D, Janaki Ramaiah M, Surendranadha Reddy J, Vijaya Bharathi E, Kashi Reddy M, Victor Prem Sagar M, Lakshminarayan Reddy T, Pushpavalli S, Pal-Bhadra M. Synthesis and apoptosis inducing ability of new anilino substituted pyrimidine sulfonamides as potential anticancer agents. Eur J Med Chem 2011; 46:5817-24. [DOI: 10.1016/j.ejmech.2011.09.039] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 09/21/2011] [Accepted: 09/21/2011] [Indexed: 11/16/2022]
|
28
|
Cole S, Bagal S, El-Kattan A, Fenner K, Hay T, Kempshall S, Lunn G, Varma M, Stupple P, Speed W. Full efficacy with no CNS side-effects: unachievable panacea or reality? DMPK considerations in design of drugs with limited brain penetration. Xenobiotica 2011; 42:11-27. [DOI: 10.3109/00498254.2011.617847] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
29
|
Min Y, He P, Wang Q, Jin X, Song B, Li L. The effects of the c-kit blocker glivec on the contractile response of urinary bladder. J Surg Res 2011; 171:e193-9. [PMID: 21962730 DOI: 10.1016/j.jss.2011.07.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 07/19/2011] [Accepted: 07/29/2011] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To investigate the effects of imatinib (Glivec) on the urinary bladder contraction and excitation induced by neurostimulation, therefore to clarify the relationships between the bladder interstitial cell of Cajal (ICC) and the neural signals. METHODS In in vivo experiments, pelvic nerves of rats were stimulated by square-wave pulses. The contractile response was recorded before and 40 min after the administration of medications (atropine, Glivec, and ketotifen). In in vitro experiments, the bladder contractile response induced by acetylcholine with or without Glivec was evaluated. The space relationship between ICC and neural fibers were observed with double-labeled fluorescence using primary antibodies (anti-c-kit and anti-vesicular acetylcholine transferase) and secondary fluorescent antibodies (Alexa 488 and Alexa 594; Molecular Probes, Eugene, OR). RESULTS Atropine and Glivec could significantly inhibit the bladder contractile response induced by the electrical stimulation in a dose-dependent manner, while ketotifen did not obviously affect bladder contractile response. In in vitro experiments, Glivec did not affect acetylcholine-induced bladder contractile response. The location of ICC in close proximity to cholinergic nerve fibers was confirmed by double-labeled fluorescence. CONCLUSIONS Bladder ICC play an important role as intermediaries in the transmission of cholinergic signals from nerve to smooth muscle cells.
Collapse
Affiliation(s)
- Yongzheng Min
- Centre of Urology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
30
|
Glekas AP, Pillarsetty NK, Punzalan B, Khan N, Smith-Jones P, Larson SM. In vivo imaging of Bcr-Abl overexpressing tumors with a radiolabeled imatinib analog as an imaging surrogate for imatinib. J Nucl Med 2011; 52:1301-7. [PMID: 21764785 DOI: 10.2967/jnumed.110.085050] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
UNLABELLED Imatinib mesylate is a tyrosine kinase inhibitor that was approved by the U.S. Food and Drug Administration in 2001 for treatment of many different stages of chronic myeloid leukemia and in 2002 for treatment of gastrointestinal stromal tumors. Imatinib is known to inhibit the dysregulated proliferation of chronic myeloid leukemia, which is associated with the Bcr-Abl kinase; in gastrointestinal stromal tumors, imatinib is known to act via c-Kit kinase inhibition. The objective of this study was to synthesize an (18)F-labeled analog of imatinib not as a primary imaging agent but rather as a tracer for in vivo drug distribution and tracer concentration that can be used as a PET imaging surrogate for imatinib. METHODS Molecular modeling studies based on the crystal structure of imatinib bound to the active site of Abl were performed for designing the fluorinated analog. A 2-fluoroethyl analog of imatinib (SKI696) was synthesized using well-established procedures. The selectivity and binding affinity of SKI696 were compared with those of imatinib in vitro. Mice bearing K562 tumor xenografts, which are known to overexpress Bcr-Abl, were imaged with (18)F-SKI696 PET. A biodistribution study was also performed on K562 tumor-bearing mice to which our radiolabeled tracer was administered. RESULTS The kinase assay verified that imatinib and SKI696 bind to the same targets with similar affinities. The feasibility of using (18)F-SKI696 as a PET agent was examined in vivo, and (18)F-SKI696 PET was shown to visualize K562 tumor xenografts in nude mice. The tumor was visible on PET 1 h after injection, with uptake of 1% of the injected dose. Biodistribution studies in K562-bearing mice were also performed, and the uptake of (18)F-SKI696 (percentage injected dose per gram) for each organ was calculated. CONCLUSION The results of the binding assay showed that SKI696 has selectivity and binding affinity comparable to imatinib. Small-animal PET of K562 tumor-bearing mice using (18)F-SKI696 as a PET agent displayed promising tumor uptake and tumor-to-nontarget contrast. Because (18)F-SKI696 has been taken up in vivo by tumors that overexpress Bcr-Abl, we are exploring a possible role for identifying tumors that will respond to imatinib before therapy.
Collapse
Affiliation(s)
- Athanasios P Glekas
- Radiochemistry Service, Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Radiolabeled small molecule protein kinase inhibitors for imaging with PET or SPECT. Molecules 2010; 15:8260-78. [PMID: 21079565 PMCID: PMC6259110 DOI: 10.3390/molecules15118260] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 11/10/2010] [Accepted: 11/11/2010] [Indexed: 01/27/2023] Open
Abstract
Imaging protein kinase expression with radiolabeled small molecule inhibitors has been actively pursued to monitor the clinical potential of targeted therapeutics and treatments as well as to determine kinase receptor density changes related to disease progression. The goal of the present review is to provide an overview of the breadth of radiolabeled small molecules that have been synthesized to target intracellular protein kinases, not only for imaging in oncology, but also for other areas of interest, particularly the central nervous system. Considerable radiotracer development has focused on imaging receptor tyrosine kinases of growth factors, protein kinases A, B and C, and glycogen synthase kinase-3ß. Design considerations, structural attributes and relevant biological results are summarized.
Collapse
|
32
|
Abstract
IMPORTANCE OF THE FIELD A concerted effort by the pharmaceutical industry over the last decade has led to the successful clinical development of protein kinase inhibitors as effective targeted therapies for certain cancers. AREAS COVERED IN THIS REVIEW This review details eight small molecule kinase inhibitors that have been approved for the treatment of cancer in either the US or Europe as of March 2010: imatinib, sorafenib, gefitinib, erlotinib, dasatinib, lapatinib, sunitinib and nilotinib. These eight compounds vary from the relatively specific inhibitor lapatinib to the more promiscuous kinase inhibitors dasatinib and sunitinib. WHAT THE READER WILL GAIN A brief discussion on the biology of each inhibitor, selectivity over other kinases and toxicity is provided. A more detailed discussion on the metabolism, drug transporters, drug-drug interactions and possible roles of metabolism in compound toxicity is provided for each compound. TAKE HOME MESSAGE The majority of the currently approved kinase inhibitors is heavily influenced by drug transporters and significantly affected by CYP3A4 inhibitors/inducers. At least three, gefitinib, erlotinib and dasatinib, are metabolized to form reactive metabolites capable of covalently-binding biomolecules.
Collapse
Affiliation(s)
- Derek R. Duckett
- Department of Molecular Therapeutics, Scripps Florida, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Michael D. Cameron
- Department of Molecular Therapeutics, Scripps Florida, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
33
|
Synthesis and biological evaluation of anilino substituted pyrimidine linked pyrrolobenzodiazepines as potential anticancer agents. Bioorg Med Chem Lett 2010; 20:5232-6. [DOI: 10.1016/j.bmcl.2010.06.147] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 06/09/2010] [Accepted: 06/30/2010] [Indexed: 12/25/2022]
|
34
|
Liu H, Xia W, Luo Y, Lu W. A novel synthesis of imatinib and its intermediates. MONATSHEFTE FUR CHEMIE 2010. [DOI: 10.1007/s00706-010-0334-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Doubrovin M, Kochetkova T, Santos E, Veach DR, Smith-Jones P, Pillarsetty N, Balatoni J, Bornmann W, Gelovani J, Larson SM. (124)I-iodopyridopyrimidinone for PET of Abl kinase-expressing tumors in vivo. J Nucl Med 2010; 51:121-9. [PMID: 20048131 DOI: 10.2967/jnumed.109.066126] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
UNLABELLED Because of the recent development of an iodopyridopyrimidinone Abl protein kinase inhibitor (PKI), (124)I-SKI-212230 ((124)I-SKI230), we investigated the feasibility of a PET-based molecular imaging method for the direct visualization of Abl kinase expression and PKI treatment. METHODS In vitro pharmacokinetic properties, including specific and nonspecific binding of (124)I-SKI230 to its Abl kinase target and interaction with other PKIs, were assessed in cell-free medium and chronic myelogenous leukemia (CML) cells overexpressing BCR-Abl (K562), in comparison with BT-474 cells that are low in Abl expression. In a xenograft tumor model, we assessed the in vivo pharmacokinetics of (124)I-SKI230 using PET and postmortem tissue sampling. We also tested a paradigm of (124)I-SKI230 PET after treatment of the animal with a dose of Abl-specific PKI for the monitoring of the tumor response. RESULTS In vitro studies confirmed that SKI230 binds to Abl kinase with nanomolar affinity, that selective uptake occurs in cell lines known to express Abl kinase, that RNAi knock-down supports specificity of cellular uptake due to Abl kinase, and that imatinib, an archetype Abl PKI, completely displaces SKI230. With SKI230, we obtained successful in vivo PET of Abl-expressing human tumors in a nude rat. We were also able to demonstrate evidence of substrate inhibition of in vivo radiotracer uptake in the xenograft tumor after treatment of the animal as a model of PKI treatment monitoring. CONCLUSION These results support the hypothesis that molecular imaging using PET will be useful for the study of in vivo pharmacodynamics of Abl PKI molecular therapy in humans.
Collapse
Affiliation(s)
- Mikhail Doubrovin
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Combined treatment strategies in gastrointestinal stromal tumors (GISTs) after imatinib and sunitinib therapy. Cancer Treat Rev 2009; 36:63-8. [PMID: 19914780 DOI: 10.1016/j.ctrv.2009.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 10/14/2009] [Accepted: 10/19/2009] [Indexed: 12/14/2022]
Abstract
Resistance to tyrosine-kinase inhibitors remains an open issue in the treatment of patients with gastrointestinal stromal tumors. The complex biology of disease in the multi-resistant setting has led a progressively growing urgency and interest in development combined or integrated therapies. This mini-review outlines the rationale for developing new combined therapeutic approaches, and describes the state of the art of the various potential strategies and the promising research perspectives.
Collapse
|
37
|
Kniess T, Bergmann R, Kuchar M, Steinbach J, Wuest F. Synthesis and radiopharmacological investigation of 3-[4'-[(18)F]fluorobenzylidene]indolin-2-one as possible tyrosine kinase inhibitor. Bioorg Med Chem 2009; 17:7732-42. [PMID: 19822433 DOI: 10.1016/j.bmc.2009.09.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 09/15/2009] [Accepted: 09/21/2009] [Indexed: 11/28/2022]
Abstract
The radiosynthesis and radiopharmacological evaluation of 3-[4'-[(18)F]fluorobenzylidene]indolin-2-one, a derivative of tyrosine kinase inhibitor SU5416, is described. The radiosynthesis was accomplished by Knoevenagel condensation of 4-[(18)F]fluorobenzaldehyde with oxindole in a remotely controlled synthesis module. The reaction conditions were optimized through screening the influence of different bases on the radiochemical yield. The radiotracer was obtained after a two-step labelling procedure in 4% decay-corrected radiochemical yield at a specific activity of 48-61GBq/micromol within 90min. The radiochemical purity after semi-preparative HPLC purification exceeded 98%. The biodistribution was studied in Wistar rats. After distribution the radiotracer was rapidly accumulated in the adrenals, liver and kidneys, however, it was cleared from these and the most other organs. Only the adipose tissue remained the activity over 60min. Unexpected high transient uptake was observed in the brain, pancreas, heart and lung. The fast clearance of 3-[4'-[(18)F]fluorobenzylidene]indolin-2-one was caused by excretion, approximately one half each was renal and biliary excreted and the other part cleared by metabolic processes. In arterial blood plasma two more polar metabolites were found by radio-HPLC. After 20min post-injection, only 12% of intact radiotracer has been detected. Consequently, in small animal PET studies with FaDu tumour bearing mice no specific uptake in the tumours could be observed.
Collapse
Affiliation(s)
- Torsten Kniess
- Institute of Radiopharmacy, Forschungszentrum Dresden-Rossendorf e.V., PO Box 510119, D-01314 Dresden, Germany.
| | | | | | | | | |
Collapse
|
38
|
Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev 2009; 35:692-706. [PMID: 19733976 DOI: 10.1016/j.ctrv.2009.08.004] [Citation(s) in RCA: 331] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 08/03/2009] [Accepted: 08/06/2009] [Indexed: 01/21/2023]
Abstract
In the recent years, eight tyrosine kinase inhibitors (TKIs) have been approved for cancer treatment and numerous are under investigation. These drugs are rationally designed to target specific tyrosine kinases that are mutated and/or over-expressed in cancer tissues. Post marketing study commitments have been made upon (accelerated) approval such as additional pharmacokinetic studies in patients with renal- or hepatic impairment, in children, additional interactions studies and studies on the relative or absolute bioavailability. Therefore, much information will emerge on the pharmacokinetic behavior of these drugs after their approval. In the present manuscript, the pharmacokinetic characteristics; absorption, distribution, metabolism and excretion (ADME), of the available TKIs are reviewed. Results from additional studies on the effect of drug transporters and drug-drug interactions have been incorporated. Overall, the TKIs reach their maximum plasma levels relatively fast; have an unknown absolute bioavailability, are extensively distributed and highly protein bound. The drugs are primarily metabolized by cytochrome P450 (CYP) 3A4 with other CYP-enzymes playing a secondary role. They are predominantly excreted with the feces and only a minor fraction is eliminated with the urine. All TKIs appear to be transported by the efflux ATP binding-cassette transports B1 and G2. Additionally these drugs can inhibit some of their own metabolizing enzymes and transporters making steady-state metabolism and drug-drug interactions both complex and unpredictable. By understanding the pharmacokinetic profile of these drugs and their similarities, factors that influence drug exposure will be better recognized and this knowledge may be used to limit sub- or supra-therapeutic drug exposure.
Collapse
|
39
|
Gardner ER, Smith NF, Figg WD, Sparreboom A. Influence of the dual ABCB1 and ABCG2 inhibitor tariquidar on the disposition of oral imatinib in mice. J Exp Clin Cancer Res 2009; 28:99. [PMID: 19591692 PMCID: PMC2717937 DOI: 10.1186/1756-9966-28-99] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Accepted: 07/10/2009] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Imatinib, a tyrosine kinase inhibitor currently approved for treatment of several malignancies, has been shown to be a substrate for multiple efflux-transporter proteins, including ABCB1 (P-glycoprotein) and ABCG2 (BCRP). The effect of inhibiting these transporters on tissue exposure to imatinib remains unclear. OBJECTIVE To assess the role of these transporters on drug disposition, 50 mg/kg imatinib was administered to Balb/C mice, 30 minutes after receiving tariquidar (10 mg/kg), an inhibitor of both ABCB1 and ABCG2, or vehicle, via oral gavage. METHODS Quantitative determination of imatinib in mouse plasma, liver and brain was performed using a newly-developed and validated liquid-chromatography-mass spectrometric method. RESULTS Exposure to imatinib was 2.2-fold higher in plasma, liver and brain in mice that received tariquidar, as compared to those that received the vehicle (P = 0.001). The peak plasma concentration did not increase substantially, suggesting that tariquidar is affecting the distribution, metabolism and/or excretion of imatinib, rather than absorption. Though tariquidar increased the absolute exposure of imatinib, the brain-to-plasma ratio of imatinib was unaffected. CONCLUSION This study suggests that intentional inhibition of ABCB1 and ABCG2 function at the blood-brain barrier is unlikely to significantly improve clinical outcome of imatinib with currently used dosing regimens.
Collapse
Affiliation(s)
- Erin R Gardner
- Clinical Pharmacology Program, SAIC-Frederick, Inc, NCI-Frederick, Frederick, Maryland, 21702, USA
| | - Nicola F Smith
- Clinical Pharmacology Program, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - William D Figg
- Clinical Pharmacology Program, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Alex Sparreboom
- Clinical Pharmacology Program, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| |
Collapse
|
40
|
Pantaleo MA, Landuzzi L, Nicoletti G, Nanni C, Boschi S, Piazzi G, Santini D, Di Battista M, Castellucci P, Lodi F, Fanti S, Lollini PL, Biasco G. Advances in preclinical therapeutics development using small animal imaging and molecular analyses: the gastrointestinal stromal tumors model. Clin Exp Med 2009; 9:199-205. [PMID: 19225718 PMCID: PMC2709232 DOI: 10.1007/s10238-009-0033-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Accepted: 01/16/2009] [Indexed: 01/13/2023]
Abstract
The large use of target therapies in the treatment of gastrointestinal stromal tumors (GISTs) highlighted the urgency to integrate new molecular imaging technologies, to develop new criteria for tumor response evaluation and to reach a more comprehensive definition of the molecular target. These aspects, which come from clinical experiences, are not considered enough in preclinical research studies which aim to evaluate the efficacy of new drugs or new combination of drugs with molecular target. We developed a xenograft animal model GIST882 using nude mice. We evaluated both the molecular and functional characterization of the tumor mass. The mutational analysis of KIT receptor of the GIST882 cell lines and tumor mass showed a mutation on exon 13 that was still present after in vivo cell growth. The glucose metabolism and cell proliferation was evaluated with a small animal PET using both FDG and FLT. The experimental development of new therapies for GIST treatment requires sophisticated animal models in order to represent the tumor molecular heterogeneity already demonstrated in the clinical setting and in order to evaluate the efficacy of the treatment also considering the inhibition of tumor metabolism, and not only considering the change in size of tumors. This approach of cancer research on GISTs is crucial and essential for innovative perspectives that could cross over to other types of cancer.
Collapse
Affiliation(s)
- M A Pantaleo
- Department of Hematology and Oncology Sciences L.A.Seragnoli, Sant'Orsola-Malpighi Hospital, University of Bologna, 40138, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Polli JW, Olson KL, Chism JP, John-Williams LS, Yeager RL, Woodard SM, Otto V, Castellino S, Demby VE. An unexpected synergist role of P-glycoprotein and breast cancer resistance protein on the central nervous system penetration of the tyrosine kinase inhibitor lapatinib (N-{3-chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methylsulfonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine; GW572016). Drug Metab Dispos 2009; 37:439-42. [PMID: 19056914 DOI: 10.1124/dmd.108.024646] [Citation(s) in RCA: 202] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lapatinib is a tyrosine kinase inhibitor approved for use in combination with capecitabine to treat advanced or metastatic breast cancers overexpressing human epidermal receptor 2 (ErbB2). This work investigated the role of P-glycoprotein (Pgp; the protein from the Mdr1a/b gene) and breast cancer resistance protein (Bcrp; the protein from the Bcrp1 gene) in modulating the central nervous system penetration of lapatinib at steady-state conditions in FVBn mice (wild-type), Mdr1a/b(-/-), Bcrp1(-/-), and Mdr1a/b(-/-)/Bcrp1(-/-) knockout mice. After an intravenous infusion of lapatinib for 24 h to a targeted steady-state plasma concentration of 700 ng/ml (0.3 mg/kg/h) or 7000 ng/ml (3 mg/kg/h), lapatinib brain-to-plasma ratios were approximately 3- to 4-fold higher in Mdr1a/b(-/-) knockout mice (ratio range from 0.09 to 0.16) compared with wild-type mice (ratio range from 0.03 to 0.04). There was no difference in the brain-to-plasma ratio in the Bcrp1(-/-) knockout mice (ratio range from 0.03 to 0.04) compared with wild-type mice. In contrast, Mdr1a/b(-/-)/Bcrp1(-/-) triple knockout mice had a 40-fold higher brain-to-plasma ratio (ratio range from 1.2 to 1.7), suggesting that Pgp and Bcrp work in concert to limit the brain-to-plasma ratio of lapatinib in mice. This finding has important potential consequences for the treatment of brain tumors in breast cancer patients treated with tyrosine kinase inhibitors as well as the basic understanding of ATP binding cassette transporters expressed in the blood-brain barrier on the central nervous system disposition of drugs.
Collapse
Affiliation(s)
- Joseph W Polli
- Preclinical Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Inc., P.O. Box 13398, Room MAI.A2213, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kil KE, Biegon A, Ding YS, Fischer A, Ferrieri RA, Kim SW, Pareto D, Schueller MJ, Fowler JS. Synthesis and PET studies of [(11)C-cyano]letrozole (Femara), an aromatase inhibitor drug. Nucl Med Biol 2009; 36:215-23. [PMID: 19217534 PMCID: PMC3161428 DOI: 10.1016/j.nucmedbio.2008.11.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 10/18/2008] [Accepted: 11/19/2008] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Aromatase, a member of the cytochrome P450 family, converts androgens such as androstenedione and testosterone into estrone and estradiol, respectively. Letrozole (1-[bis-(4-cyanophenyl)methyl]-1H-1,2,4-triazole; Femara) is a high-affinity aromatase inhibitor (K(i)=11.5 nM) that has Food and Drug Administration approval for breast cancer treatment. Here we report the synthesis of carbon-11-labeled letrozole and its assessment as a radiotracer for brain aromatase in the baboon. METHODS Letrozole and its precursor (4-[(4-bromophenyl)-1H-1,2,4-triazol-1-ylmethyl]benzonitrile) were prepared in a two-step synthesis from 4-cyanobenzyl bromide and 4-bromobenzyl bromide, respectively. The [(11)C]cyano group was introduced via tetrakis(triphenylphosphine)palladium(0)-catalyzed coupling of [(11)C]cyanide with the bromo precursor. Positron emission tomography (PET) studies in the baboon brain were carried out to assess regional distribution and kinetics, reproducibility of repeated measures and saturability. Log D, the free fraction of letrozole in plasma and the [(11)C-cyano]letrozole fraction in arterial plasma were also measured. RESULTS [(11)C-cyano]Letrozole was synthesized in 60 min with a radiochemical yield of 79-80%, with a radiochemical purity greater than 98% and a specific activity of 4.16+/-2.21 Ci/mumol at the end of bombardment (n=4). PET studies in the baboon revealed initial rapid and high uptake and initial rapid clearance, followed by slow clearance of carbon-11 from the brain, with no difference between brain regions. Brain kinetics was not affected by coinjection of unlabeled letrozole (0.1 mg/kg). The free fraction of letrozole in plasma was 48.9%, and log D was 1.84. CONCLUSION [(11)C-cyano]Letrozole is readily synthesized via a palladium-catalyzed coupling reaction with [(11)C]cyanide. Although it is unsuitable as a PET radiotracer for brain aromatase, as revealed by the absence of regional specificity and saturability in brain regions such as amygdala, which are known to contain aromatase, it may be useful in measuring letrozole distribution and pharmacokinetics in the brain and peripheral organs.
Collapse
Affiliation(s)
- Kun-Eek Kil
- Medical Department, Brookhaven National Laboratory, Upton, NY 11973
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794
| | - Anat Biegon
- Medical Department, Brookhaven National Laboratory, Upton, NY 11973
| | - Yu-Shin Ding
- Department of Radiology, Yale University School of Medicine, New Haven, CT 06520-8048
| | - Andre Fischer
- Johannes-Gutenberg Universität Mainz, Institut für Organische Chemie, Duesbergweg 10-14, Mainz 55128 Germany
| | | | - Sung Won Kim
- Medical Department, Brookhaven National Laboratory, Upton, NY 11973
| | - Deborah Pareto
- Medical Department, Brookhaven National Laboratory, Upton, NY 11973
| | | | - Joanna S. Fowler
- Medical Department, Brookhaven National Laboratory, Upton, NY 11973
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794
| |
Collapse
|
43
|
Marantidou A, Carpentier A. Nouvelles thérapeutiques ciblées dans les glioblastomes. ONCOLOGIE 2009. [DOI: 10.1007/s10269-008-1035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
44
|
|
45
|
Ivanov AS, Shishkov SV. Synthesis of imatinib: a convergent approach revisited. MONATSHEFTE FUR CHEMIE 2009. [DOI: 10.1007/s00706-008-0105-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
46
|
Eckelman WC, Reba RC, Kelloff GJ. Targeted imaging: an important biomarker for understanding disease progression in the era of personalized medicine. Drug Discov Today 2008; 13:748-59. [PMID: 18617011 DOI: 10.1016/j.drudis.2008.05.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 04/30/2008] [Accepted: 05/22/2008] [Indexed: 11/16/2022]
Abstract
The key to applying targeted imaging to personalized medicine is the choice of the right radiolabeled probe for the right target for the right disease following the lead of pharmaceutical development. The imaging approach differs depending on whether the target is a single disease control point (e.g. a specific receptor or transport protein linked to the mechanistic activity of a drug) or a general disease control point applicable to a number of treatment paradigms (e.g. proliferation, angiogenesis, inflammation). But in either case, the number of control points must be small given the time constraints on molecular imaging procedures in the clinic. Regardless of the choice, the radiotracer must be validated as binding to the target with the appropriate pharmacokinetics and pharmacodynamics for effective external imaging. Such an imaging agent developed in concert with drug development has a built in synergy that will accelerate the drug development process, targeted imaging and personalized medicine as well.
Collapse
|
47
|
Reaction of 1-(3,5-Bis(trifluoromethyl)phenyl)-3-(4-(Pyridin-3-yl)pyrimidin-2-yl) urea with Methyl Iodide and X-ray Crystallographic Structure of Its Derivative. B KOREAN CHEM SOC 2008. [DOI: 10.5012/bkcs.2008.29.7.1421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Le Guilloux J, Carpentier AF. [New therapeutic approaches in glioblastomas]. Rev Neurol (Paris) 2008; 164:554-9. [PMID: 18565354 DOI: 10.1016/j.neurol.2008.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 03/25/2008] [Indexed: 01/29/2023]
Abstract
Current treatment of glioblastomas relies on surgical resection, radiotherapy and chemotherapy. However, the efficacy of these therapeutics is still limited and new therapeutic approaches based on the understanding of brain tumor biology are emerging. High expression of the EGF receptor by tumor cells, activation of the PI3K/Akt and the Ras/Raf pathways represent interesting targets for new selective drugs under development. The most promising drugs are currently antiangiogenic agents. This article reviews these emerging therapies currently under clinical trials in glioblastomas.
Collapse
Affiliation(s)
- J Le Guilloux
- Service de neurologie de l'hôpital Avicenne, Assistance publique-Hôpitaux de Paris, 125, route de Stalingrad, 75013 Bobigny, France
| | | |
Collapse
|
49
|
El-Deeb IM, Ryu JC, Lee SH. Synthesis of new N-arylpyrimidin-2-amine derivatives using a palladium catalyst. Molecules 2008; 13:818-30. [PMID: 18463584 PMCID: PMC6245071 DOI: 10.3390/molecules13040818] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 04/08/2008] [Accepted: 04/08/2007] [Indexed: 11/19/2022] Open
Abstract
New N-aryl-4-(pyridin-3-yl)pyrimidin-2-amine derivatives were synthesized from the corresponding amines, applying optimized Buchwald-Hartwig amination conditions using dichlorobis(triphenylphosphine)Pd(II), xantphos and sodium tert-butoxide in refluxing toluene under a nitrogen atmosphere. The target N-aryl derivatives were obtained in moderate to good yields ranging from 27% to 82%. The procedure described could be widely employed for the preparation of new heterocyclic compounds. The structures of the new compounds were confirmed by FT-NMR, FT-IR and elemental analysis.
Collapse
Affiliation(s)
- Ibrahim Mustafa El-Deeb
- Department of Biomolecular Science, University of Science and Technology, Daejon, Korea; E-mail:
| | - Jae Chun Ryu
- Life Sciences Research Division, Korea Institute of Science & Technology, P.O. Box 131, Cheongryang, Seoul 130-650, Korea; E-mail:
| | - So Ha Lee
- Department of Biomolecular Science, University of Science and Technology, Daejon, Korea; E-mail:
- Life Sciences Research Division, Korea Institute of Science & Technology, P.O. Box 131, Cheongryang, Seoul 130-650, Korea; E-mail:
- Author to whom correspondence should be addressed; E-mail: ; Tel: (+82) 2 9586834
| |
Collapse
|
50
|
Polli JW, Humphreys JE, Harmon KA, Castellino S, O'Mara MJ, Olson KL, John-Williams LS, Koch KM, Serabjit-Singh CJ. The role of efflux and uptake transporters in [N-{3-chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methylsulfonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine (GW572016, lapatinib) disposition and drug interactions. Drug Metab Dispos 2008; 36:695-701. [PMID: 18216274 DOI: 10.1124/dmd.107.018374] [Citation(s) in RCA: 192] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Lapatinib [N-{3-chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methylsulfonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, GW572016, Tykerb] is a tyrosine kinase inhibitor approved for use in combination with capecitabine to treat advanced or metastatic breast cancers overexpressing HER2 (ErbB2). In this work we investigated the role of efflux and uptake transporters in lapatinib disposition and drug interactions. In vitro studies evaluated whether lapatinib is a substrate for efflux transporters or an inhibitor of efflux/uptake transporters. In vivo studies included whole-body autoradiography and an evaluation of the role of efflux transporters on the intestinal absorption and brain penetration of lapatinib using chemical or genetic knockout animals. Lapatinib is a substrate for the efflux transporters P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP). Furthermore, lapatinib is an inhibitor (IC(50) values 0.025-5 muM) of Pgp, BCRP, and organic anion transporting polypeptide 1B1 (a hepatic uptake transporter). In contrast, lapatinib yielded little inhibition on renal transporters (organic anion transporters, organic cation transporters, and uric acid transporter). In vivo studies demonstrated that brain concentrations of lapatinib were low and influenced by efflux transporters at the blood-brain barrier. In contrast, systemic exposure of lapatinib after oral dosing was unchanged when efflux by Pgp and BCRP was absent from the gastrointestinal tract. These in vitro and in vivo preclinical investigations provide a mechanistic basis for elucidating clinical drug interactions.
Collapse
Affiliation(s)
- Joseph W Polli
- Preclinical Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Inc., P.O. Box 13398, Room MAI.A2213, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|