1
|
Garrido-García P, Quirós I, Milán-Rois P, Ortega-Gutiérrez S, Martín-Fontecha M, Campos LA, Somoza Á, Fernández I, Rigotti T, Tortosa M. Enantioselective photocatalytic synthesis of bicyclo[2.1.1]hexanes as ortho-disubstituted benzene bioisosteres with improved biological activity. Nat Chem 2025; 17:734-745. [PMID: 40000889 DOI: 10.1038/s41557-025-01746-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/17/2025] [Indexed: 02/27/2025]
Abstract
1,5-Disubstituted bicyclo[2.1.1]hexanes are bridged scaffolds with well-defined exit vectors that are becoming increasingly popular building blocks in medicinal chemistry because they are saturated bioisosteres of ortho-substituted phenyl rings. Here we have developed a Lewis-acid-catalysed [2 + 2] photocycloaddition to obtain these motifs as enantioenriched scaffolds, providing an efficient approach for their incorporation in a variety of drug analogues. Retention of the biological activity of the bicyclo[2.1.1]hexane-containing analogues in the specific proteins targeted by the original drugs has confirmed the suitability of this moiety to serve as a bioisostere of ortho-substituted phenyl rings. Moreover, we have studied the potential of the different enantiomers of the drug analogues to selectively induce cytotoxicity in a panel of tumour cell lines, observing markedly differential effects for the two enantiomers and a substantial improvement over the corresponding sp2-based drugs. This showcases that the control of the absolute configuration and tridimensionality of the drug analogue has a large impact on its biological properties.
Collapse
Affiliation(s)
- Pablo Garrido-García
- Organic Chemistry Department, Faculty of Science, Autonomous University of Madrid, Madrid, Spain
| | - Irene Quirós
- Organic Chemistry Department, Faculty of Science, Autonomous University of Madrid, Madrid, Spain
| | | | - Silvia Ortega-Gutiérrez
- Organic Chemistry Department, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, Spain
| | - Mar Martín-Fontecha
- Organic Chemistry Department, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | | | | | - Israel Fernández
- Organic Chemistry Department, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, Spain
- Center of Innovation in Advanced Chemistry (ORFEO-CINQA), Madrid, Spain
| | - Thomas Rigotti
- Organic Chemistry Department, Faculty of Science, Autonomous University of Madrid, Madrid, Spain.
| | - Mariola Tortosa
- Organic Chemistry Department, Faculty of Science, Autonomous University of Madrid, Madrid, Spain.
- Center of Innovation in Advanced Chemistry (ORFEO-CINQA), Madrid, Spain.
- Institute for Advanced Research in Chemical Sciences (IAdChem), Autonomous University of Madrid, Madrid, Spain.
| |
Collapse
|
2
|
Xiang Z, Deng X, He W, Yang Q, Ni L, Dehghan Shasaltaneh M, Maghsoudloo M, Yang G, Wu J, Imani S, Wen Q. Treatment of malignant pleural effusion in non-small cell lung cancer with VEGF-directed therapy. Ann Med 2022; 54:1357-1371. [PMID: 35543207 PMCID: PMC9103356 DOI: 10.1080/07853890.2022.2071977] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is a critical regulator of malignant pleural effusion (MPE) in non-small-cell lung cancer (NSCLC). Bevacizumab (BEV) and apatinib (APA) are novel VEGF blockers that inhibit lung cancer cell proliferation and the development of pleural effusion. METHODS In this study, we established Lewis lung cancer (LLC) xenograft mouse models to compare the therapeutic effect of APA and BEV in combination with cisplatin (CDDP) against MPE. The anti-tumour and anti-angiogenic effects of this combination therapy were evaluated by 18F-FDG PET/CT imaging, TUNEL assay and Immunohistochemistry. RESULTS The triple drug combination significantly prolonged the overall survival of the tumour-bearing mice by reducing MPE and glucose metabolism and was more effective in lowering VEGF/soluble VEGFR-2 levels in the serum and pleural exudates compared to either of the monotherapies. Furthermore, CDDP + APA + BEV promoted in vivo apoptosis and decreased microvessel density. CONCLUSIONS Mechanistically, LLC-induced MPE was inhibited by targeting the VEGF-MEK/ERK pathways. Further studies are needed to establish the synergistic therapeutic effect of these drugs in NSCLC patients with MPE.KEY MESSAGESCombined treatment of MPE with apatinib, bevacizumab and cisplatin can prolong the survival time of mice, reduce the content of MPE, decrease the SUVmax of thoracic tumour tissue, down-regulate the content of VEGF and sVEGFR-2 in serum and pleural fluid, and promote the apoptosis of tumour cells. Angiogenesis and MPE formation can be inhibited by down-regulation of HIF-1α, VEGF, VEGFR-2, MEK1 and MMP-2 molecular signalling pathway proteins.
Collapse
Affiliation(s)
- Zhangqiang Xiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Phase 1 Clinical Trial Center, Deyang People's Hospital, Deyang, China
| | - Xiangyu Deng
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenfeng He
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qian Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Laichao Ni
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | | | - Mazaher Maghsoudloo
- Laboratory of Systems Biology and Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.,Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Gang Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Oncology, Anyue Hospital of Traditional Chinese Medicine, Second Ziyang Hospital of Traditional Chinese Medicine, Ziyang, China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China. The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
3
|
Li D, Patel CB, Xu G, Iagaru A, Zhu Z, Zhang L, Cheng Z. Visualization of Diagnostic and Therapeutic Targets in Glioma With Molecular Imaging. Front Immunol 2020; 11:592389. [PMID: 33193439 PMCID: PMC7662122 DOI: 10.3389/fimmu.2020.592389] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/08/2020] [Indexed: 02/04/2023] Open
Abstract
Gliomas, particularly high-grade gliomas including glioblastoma (GBM), represent the most common and malignant types of primary brain cancer in adults, and carry a poor prognosis. GBM has been classified into distinct subgroups over the years based on cellular morphology, clinical characteristics, biomarkers, and neuroimaging findings. Based on these classifications, differences in therapeutic response and patient outcomes have been established. Recently, the identification of complex molecular signatures of GBM has led to the development of diverse targeted therapeutic regimens and translation into multiple clinical trials. Chemical-, peptide-, antibody-, and nanoparticle-based probes have been designed to target specific molecules in gliomas and then be visualized with multimodality molecular imaging (MI) techniques including positron emission tomography (PET), single-photon emission computed tomography (SPECT), near-infrared fluorescence (NIRF), bioluminescence imaging (BLI), and magnetic resonance imaging (MRI). Thus, multiple molecules of interest can now be noninvasively imaged to guide targeted therapies with a potential survival benefit. Here, we review developments in molecular-targeted diagnosis and therapy in glioma, MI of these targets, and MI monitoring of treatment response, with a focus on the biological mechanisms of these advanced molecular probes. MI probes have the potential to noninvasively demonstrate the pathophysiologic features of glioma for diagnostic, treatment, and response assessment considerations for various targeted therapies, including immunotherapy. However, most MI tracers are in preclinical development, with only integrin αVβ3 and isocitrate dehydrogenase (IDH)-mutant MI tracers having been translated to patients. Expanded international collaborations would accelerate translational research in the field of glioma MI.
Collapse
Affiliation(s)
- Deling Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China
| | - Chirag B. Patel
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
- Division of Neuro-Oncology, Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, United States
| | - Guofan Xu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Andrei Iagaru
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
4
|
Wang M, Zeng Q, Li Y, Imani S, Xie D, Li Y, Han Y, Fan J. Bevacizumab combined with apatinib enhances antitumor and anti-angiogenesis effects in a lung cancer model in vitro and in vivo. J Drug Target 2020; 28:961-969. [PMID: 32374627 DOI: 10.1080/1061186x.2020.1764963] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Angiogenesis is involved in the proliferation and metastasis of solid tumours; hence, it is an attractive therapeutic target. However, most patients who undergo anti-angiogenic drug treatment do not achieve complete tumour regression, resulting in drug resistance. The objective of this research is to explore the therapeutic effect of combining bevacizumab (Bev), an anti-vascular endothelial growth factor (VEGF)-A antibody, with apatinib (Apa), a VEGR receptor (VEGFR)-2-targeting tyrosine kinase inhibitor, in non-small cell lung cancer (NSCLC). In vitro, we assessed the influence which Bev + Apa treatment exerts upon the proliferation as well as apoptosis of Lewis lung carcinoma (LLC) cells in virtue of the 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide as assay as well as Annexin V staining, respectively. For in vivo assessment, we established a tumour-bearing mouse model with LLC cells and investigated the anti-angiogenic and antitumor effects of Bev + Apa by 18F-FDG PET/CT imaging, immunohistochemistry and TUNEL staining. Bev + Apa treatment significantly inhibited LLC cell growth and proliferation in a larger scale compared to therapy of either of the only agent. Bev + Apa inhibited tumour growth and extended the median survival time of tumour-bearing mice. Mechanistically, Bev + Apa reduced angiogenesis by inhibiting VEGF and VEGFR-2 expression and reducing glucose metabolism in tumour tissues. Thus, Bev and Apa inhibited tumour angiogenesis synergistically, indicating their potential clinical utility for NSCLC treatment.
Collapse
Affiliation(s)
- Mingting Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, P.R. China
| | - Qin Zeng
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, P.R. China
| | - Yuan Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, P.R. China
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, P.R. China
| | - Danna Xie
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, P.R. China
| | - Yinghua Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, P.R. China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, P.R. China
| | - Juan Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, P.R. China
| |
Collapse
|
5
|
Iommelli F, De Rosa V, Terlizzi C, Fonti R, Del Vecchio S. Preclinical Imaging in Targeted Cancer Therapies. Semin Nucl Med 2019; 49:369-381. [PMID: 31470932 DOI: 10.1053/j.semnuclmed.2019.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Preclinical imaging with radiolabeled probes can provide noninvasive tools to test the efficacy of targeted agents in tumors harboring specific genetic alterations and to identify imaging parameters that can be used as pharmacodynamics markers in cancer patients. The present review will primarily focus on preclinical imaging studies that can accelerate the clinical approval of targeted agents and promote the development of imaging biomarkers for clinical applications. Since only subgroups of patients may benefit from treatment with targeted anticancer agents, the identification of a patient population expressing the target is of primary importance for the success of clinical trials. Preclinical imaging studies tested the ability of new radiolabeled compounds to recognize mutant, amplified, or overexpressed targets and some of these tracers were transferred to the clinical setting. More common tracers such as 18F-Fluorothymidine and 18F-Fluorodeoxyglucose were employed in animal models to test the inhibition of the target and downstream pathways through the evaluation of early changes of proliferation and glucose metabolism allowing the identification of sensitive and resistant tumors. Furthermore, since the majority of patients treated with targeted anticancer agents will invariably develop resistance, preclinical imaging studies were performed to test the efficacy of reversal agents to overcome resistance. These studies provided consistent evidence that imaging with radiolabeled probes can monitor the reversal of drug resistance by newly designed alternative compounds. Finally, despite many difficulties and challenges, preclinical imaging studies targeting the expression of immune checkpoints proved the principle that it is feasible to select patients for immunotherapy based on imaging findings. In conclusion, preclinical imaging can be considered as an integral part of the complex translational process that moves a newly developed targeted agent from laboratory to clinical application intervening in all clinically relevant steps including patient selection, early monitoring of drug effects and reversal of drug resistance.
Collapse
Affiliation(s)
- Francesca Iommelli
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Viviana De Rosa
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Cristina Terlizzi
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Rosa Fonti
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
6
|
Provost C, Rozenblum-Beddok L, Nataf V, Merabtene F, Prignon A, Talbot JN. [ 68Ga]RGD Versus [ 18F]FDG PET Imaging in Monitoring Treatment Response of a Mouse Model of Human Glioblastoma Tumor with Bevacizumab and/or Temozolomide. Mol Imaging Biol 2019; 21:297-305. [PMID: 29948641 DOI: 10.1007/s11307-018-1224-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE The aim of this study was to evaluate positron emission tomography (PET) imaging with [68Ga]NODAGA-c(RGDfK) ([68Ga]RGD), in comparison with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG), for early monitoring of the efficacy of an antiangiogenic agent associated or not with chemotherapy, in a mouse model of glioblastoma (GB). PROCEDURES Mice bearing U87MG human GB cells line were parted into five groups of five mice each. One group was imaged at baseline before the treatment phase; another group was treated with bevacizumab (BVZ), another group with temozolomide (TMZ), another group with both agents, and the last one was the control group. Tumors growth and biological properties were evaluated by caliper measurements and PET imaging at three time points (baseline, during treatment t1 = 4-6 days and t2 = 10-12 days). At the end of the study, tumors were counted and analyzed by immunohistochemistry (CD31 to evaluate microvessel density). RESULTS The tumor volume assessed by caliper measurements was significantly greater at t1 in the control group than in the TMZ + BVZ-treated group or in the BVZ-treated group. At t2, tumor volume of all treated groups was significantly smaller than that of the control group. [18F]FDG PET failed to reflect this efficacy of treatment. In contrast, at t1, the [68Ga]RGD tumor uptake was concordant with tumor growth in controls and in treated groups. At t2, a significant increase in tumor uptake of [68Ga]RGD vs. t1 was only observed in the TMZ-treated group, reflecting a lack of angiogenesis inhibition, whereas TMZ + BVZ resulted in a dramatic tumor arrest, reduction in microvessel density and stable tumor [68Ga]RGD uptake. CONCLUSIONS [68Ga]RGD is a useful PET agent for in vivo angiogenesis imaging and can be useful for monitoring antiangiogenic treatment associated or not with chemotherapy.
Collapse
Affiliation(s)
- Claire Provost
- Laboratoire d'Imagerie Moléculaire Positonique (LIMP), UMS 28, UPMC - Sorbonne Universités, Paris, France.
| | - Laura Rozenblum-Beddok
- Laboratoire d'Imagerie Moléculaire Positonique (LIMP), UMS 28, UPMC - Sorbonne Universités, Paris, France.,Service de Médecine Nucléaire et Radiopharmacie, Hôpital Tenon, AP-HP, Paris, France
| | - Valérie Nataf
- Laboratoire d'Imagerie Moléculaire Positonique (LIMP), UMS 28, UPMC - Sorbonne Universités, Paris, France.,Service de Médecine Nucléaire et Radiopharmacie, Hôpital Tenon, AP-HP, Paris, France
| | - Fatiha Merabtene
- Plateforme d'Histomorphologie Service d'Anatomie Pathologique, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Aurélie Prignon
- Laboratoire d'Imagerie Moléculaire Positonique (LIMP), UMS 28, UPMC - Sorbonne Universités, Paris, France
| | - Jean-Noël Talbot
- Laboratoire d'Imagerie Moléculaire Positonique (LIMP), UMS 28, UPMC - Sorbonne Universités, Paris, France.,Service de Médecine Nucléaire et Radiopharmacie, Hôpital Tenon, AP-HP, Paris, France
| |
Collapse
|
7
|
Andriu A, Crockett J, Dall'Angelo S, Piras M, Zanda M, Fleming IN. Binding of α vβ 3 Integrin-Specific Radiotracers Is Modulated by Both Integrin Expression Level and Activation Status. Mol Imaging Biol 2018; 20:27-36. [PMID: 28695371 PMCID: PMC5775384 DOI: 10.1007/s11307-017-1100-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Molecular imaging of αvβ3 integrin has exhibited real potential to guide the appropriate use of anti-angiogenic therapies. However, an incomplete understanding of the factors that influence binding of αvβ3 integrin-specific radiotracers currently limits their use for assessing response to therapy in cancer patients. This study identifies two fundamental factors that modulate uptake of these radiotracers. Procedures Experiments were performed in prostate cancer (PC3) and glioblastoma (U87MG) cells, which differentially express αvβ3 integrin. αvβ3 integrin-specific radiotracers were used to investigate the effect of manipulating αvβ3 integrin expression or activation in cellular binding assays. β3 integrin and αvβ3 integrin expression were measured by western blotting and flow cytometry, respectively. The effect of select pharmacological inhibitors on αvβ3 integrin activation and expression was also determined. RESULTS Radiotracer binding was proportional to αvβ3 integrin expression when it was decreased (β3 knock-down cells) or increased, either using pharmacological inhibitors of cell signalling or by culturing cells for different times. Studies with both small molecule and arginine-glycine-aspartic acid (RGD)-based radiotracers revealed increased radiotracer binding after activation of αvβ3 integrin with Mn2+ or talin head domain. Moreover, inhibition of fundamental signalling pathways (mitogen-activated protein kinase kinase (MEK), Src and VEGFR2) decreased radiotracer binding, reflecting reduced αvβ3 integrin activity. CONCLUSION Binding of small molecule ligands and radiolabelled RGD peptides is modulated by expression and activation status of αvβ3 integrin. αvβ3 integrin-specific radiotracers can provide otherwise inaccessible information of the effect of signalling pathways on αvβ3 integrin. This has significant implications for assessing response to anti-angiogenic therapies in clinical studies.
Collapse
Affiliation(s)
- Alexandra Andriu
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Julie Crockett
- Arthritis and Musculoskeletal Medicine Research Programme, Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Sergio Dall'Angelo
- Kosterlitz Centre for Therapeutics, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, AB25 2ZD, UK
| | - Monica Piras
- Kosterlitz Centre for Therapeutics, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, AB25 2ZD, UK
| | - Matteo Zanda
- Kosterlitz Centre for Therapeutics, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, AB25 2ZD, UK
| | - Ian N Fleming
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
8
|
Comparison and evaluation of two RGD peptides labelled with 68Ga or 18F for PET imaging of angiogenesis in animal models of human glioblastoma or lung carcinoma. Oncotarget 2018; 9:19307-19316. [PMID: 29721204 PMCID: PMC5922398 DOI: 10.18632/oncotarget.25028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 03/19/2018] [Indexed: 12/25/2022] Open
Abstract
The aim of this study was to evaluate two RGD radiotracers radiolabelled with fluorine-18 or gallium-68, in detecting angiogenesis in grafted human tumours and monitoring their treatment with the anti-angiogenic agent bevacizumab. Sixteen mice bearing an U87MG tumour in one flank and a contralateral A549 tumour were treated with intravenous injections of bevacizumab twice a week for 3 weeks. PET images with 18F-RGD-K5 and 68Ga-RGD were acquired before treatment (baseline), after three bevacizumab injections (t1) and after seven bevacizumab injections (t2). In A549 tumours, the treatment stopped the tumour growth, with a tumour volume measured by calliper remaining between 0.28 and 0.40 cm3. The decrease in tumour uptake of both RGD tracers was non-significant. Therefore it was not possible to predict this efficacy on tumour growth based on RGD PET results, whereas ex vivo measurements showed a significantly lower tumour uptake of both tracers in mice sacrificed at t2 vs. at baseline. In U87MG tumours, the uptake measured on PET decreased during treatment, reflecting the partial therapeutic effect observed on tumour volume, consisting in a decrease in the slope of tumour growth. Using 18F-RGD-K5, this decrease in tumour SUVmax became significant at t1, whereas it was also observed with the 68Ga-RGD tracer, but only at t2. 18F-RGD-K5 appeared more efficient than 68Ga-RGD in the visualisation and follow-up of U87MG tumours. The comparison of those results with those of immunohistochemistry at baseline and at t2 favoured the hypothesis that tumour RGD uptake reflects other cancer properties than just its angiogenic capacity.
Collapse
|
9
|
Abbasi AZ, Gordijo CR, Amini MA, Maeda A, Rauth AM, DaCosta RS, Wu XY. Hybrid Manganese Dioxide Nanoparticles Potentiate Radiation Therapy by Modulating Tumor Hypoxia. Cancer Res 2016; 76:6643-6656. [PMID: 27758881 DOI: 10.1158/0008-5472.can-15-3475] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 11/16/2022]
Abstract
Hypoxia in the tumor microenvironment (TME) mediates resistance to radiotherapy and contributes to poor prognosis in patients receiving radiotherapy. Here we report the design of clinically suitable formulations of hybrid manganese dioxide (MnO2) nanoparticles (MDNP) using biocompatible materials to reoxygenate the TME by reacting with endogenous H2O2 MDNP containing hydrophilic terpolymer-protein-MnO2 or hydrophobic polymer-lipid-MnO2 provided different oxygen generation rates in the TME relevant to different clinical settings. In highly hypoxic murine or human xenograft breast tumor models, we found that administering either MDNP formulation before radiotherapy modulated tumor hypoxia and increased radiotherapy efficacy, acting to reduce tumor growth, VEGF expression, and vascular density. MDNP treatment also increased apoptosis and DNA double strand breaks, increasing median host survival 3- to 5-fold. Notably, in the murine model, approximately 40% of tumor-bearing mice were tumor-free after a single treatment with MDNPs plus radiotherapy at a 2.5-fold lower dose than required to achieve the same curative treatment without MDNPs. Overall, our findings offer a preclinical proof of concept for the use of MDNP formulations as effective radiotherapy adjuvants. Cancer Res; 76(22); 6643-56. ©2016 AACR.
Collapse
Affiliation(s)
- Azhar Z Abbasi
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Claudia R Gordijo
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad Ali Amini
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Azusa Maeda
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Andrew M Rauth
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Ralph S DaCosta
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada.,Techna Institute, University Health Network, Toronto Ontario, Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Bao X, Wang MW, Luo JM, Wang SY, Zhang YP, Zhang YJ. Optimization of Early Response Monitoring and Prediction of Cancer Antiangiogenesis Therapy via Noninvasive PET Molecular Imaging Strategies of Multifactorial Bioparameters. Theranostics 2016; 6:2084-2098. [PMID: 27698942 PMCID: PMC5039682 DOI: 10.7150/thno.13917] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 07/30/2016] [Indexed: 12/13/2022] Open
Abstract
Objective: Antiangiogenesis therapy (AAT) has provided substantial benefits regarding improved outcomes and survival for suitable patients in clinical settings. Therefore, the early definition of therapeutic effects is urgently needed to guide cancer AAT. We aimed to optimize the early response monitoring and prediction of AAT efficacy, as indicated by the multi-targeted anti-angiogenic drug sunitinib in U87MG tumors, using noninvasive positron emission computed tomography (PET) molecular imaging strategies of multifactorial bioparameters. Methods: U87MG tumor mice were treated via intragastric injections of sunitinib (80 mg/kg) or vehicle for 7 consecutive days. Longitudinal MicroPET/CT scans with 18F-FDG, 18F-FMISO, 18F-ML-10 and 18F-Alfatide II were acquired to quantitatively measure metabolism, hypoxia, apoptosis and angiogenesis on days 0, 1, 3, 7 and 13 following therapy initiation. Tumor tissues from a dedicated group of mice were collected for immunohistochemical (IHC) analysis of key biomarkers (Glut-1, CA-IX, TUNEL, ανβ3 and CD31) at the time points of PET imaging. The tumor sizes and mouse weights were measured throughout the study. The tumor uptake (ID%/gmax), the ratios of the tumor/muscle (T/M) for each probe, and the tumor growth ratios (TGR) were calculated and used for statistical analyses of the differences and correlations. Results: Sunitinib successfully inhibited U87MG tumor growth with significant differences in the tumor size from day 9 after sunitinib treatment compared with the control group (P < 0.01). The uptakes of 18F-FMISO (reduced hypoxia), 18F-ML-10 (increased apoptosis) and 18F-Alfatide II (decreased angiogenesis) in the tumor lesions significantly changed during the early stage (days 1 to 3) of sunitinib treatment; however, the uptake of 18F-FDG (increased glucose metabolism) was significantly different during the late stage. The PET imaging data of each probe were all confirmed via ex vivo IHC of the relevant biomarkers. Notably, the PET imaging of 18F-Alfatide II and 18F-FMISO was significantly correlated (all P < 0.05) with TGR, whereas the imaging of 18F-FDG and 18F-ML-10 was not significantly correlated with TGR. Conclusion: Based on the tumor uptake of the PET probes and their correlations with MVD and TGR, 18F-Alfatide II PET may not only monitor the early response but also precisely predict the therapeutic efficacy of the multi-targeted, anti-angiogenic drug sunitinib in U87MG tumors. In conclusion, it is feasible to optimize the early response monitoring and efficacy prediction of cancer AAT using noninvasive PET molecular imaging strategies of multifactorial bioparameters, such as angiogenesis imaging with 18F-Alfatide II, which represents an RGD-based probe.
Collapse
|
11
|
Withofs N, Hustinx R. Integrin αvβ3 and RGD-based radiopharmaceuticals. MEDECINE NUCLEAIRE-IMAGERIE FONCTIONNELLE ET METABOLIQUE 2016. [DOI: 10.1016/j.mednuc.2015.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
12
|
Kong WT, Yuan HX, Cai H, Wang WP, Tang Y, Zhang XL. Early treatment response to sorafenib for rabbit VX2 orthotic liver tumors: evaluation by quantitative contrast-enhanced ultrasound. Tumour Biol 2015; 36:2593-2599. [PMID: 25448880 DOI: 10.1007/s13277-014-2877-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/18/2014] [Indexed: 11/27/2022] Open
Abstract
The aim of our study was to investigate the application of contrast-enhanced ultrasound (CEUS) and its quantification analysis for the prediction of early treatment response of sorafenib on rabbit VX2 liver tumor model. Rabbits were implanted VX2 tumor mass to establish a liver tumor model. Fourteen days after tumor implantation, rabbits presented with single liver tumor were randomly divided into two groups. Rabbits in treatment group were given by gavage once a day for 14 days with sorafenib suspension at a dose of 30 mg/kg, whereas rabbits in control group were given saline by gavage of the same volume. CEUS was performed before treatment and 3, 7, 14 days after treatment for the analysis of tumor size, enhancement pattern, and necrosis range. The time intensity curve (TIC) was used to obtain quantitative parameters of enhancement patterns. Before sorafenib administration, tumor volumes ranged from 0.24 to 0.75 cm(3) (mean 0.49 ± 0.18 cm(3)) in treatment group and 0.24 to 0.44 cm(3) (mean 0.30 ± 0.12 cm(3)) in control group. The dynamic enhancement patterns of tumors were homogeneous hyper-enhancement (n = 8), heterogeneous hyper-enhancement (n = 4), and peripheral rim-like enhancement (n = 2). All tumors of the treatment group presented with peripheral rim-like enhancement with large necrotic area after sorafenib administration, whereas tumors of the control group showed heterogeneous hyper-enhancement (n = 5) and peripheral rim-like enhancement (n = 2). There was a significant difference in area under the curve (AUC) before and after sorafenib treatment (P = 0.045). CEUS may be of value in the evaluation of early therapeutic response after sorafenib administration.
Collapse
Affiliation(s)
- Wen-Tao Kong
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | | | | | | | | | | |
Collapse
|
13
|
García-Figueiras R, Padhani AR, Beer AJ, Baleato-González S, Vilanova JC, Luna A, Oleaga L, Gómez-Caamaño A, Koh DM. Imaging of Tumor Angiogenesis for Radiologists--Part 2: Clinical Utility. Curr Probl Diagn Radiol 2015; 44:425-36. [PMID: 25863438 DOI: 10.1067/j.cpradiol.2015.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 02/24/2015] [Accepted: 02/28/2015] [Indexed: 12/26/2022]
Abstract
Angiogenesis is a key cancer hallmark involved in tumor growth and metastasis development. Angiogenesis and tumor microenvironment significantly influence the response of tumors to therapies. Imaging techniques have changed our understanding of the process of angiogenesis, the resulting vascular performance, and the tumor microenvironment. This article reviews the status and potential clinical value of the imaging modalities used to assess the status of tumor vasculature in vivo, before, during, and after treatment.
Collapse
Affiliation(s)
- Roberto García-Figueiras
- Department of Radiology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.
| | - Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, England, UK
| | - Ambros J Beer
- Klinik für Nuklearmedizin, Universitätsklinikum Ulm; Ulm, Germany
| | - Sandra Baleato-González
- Department of Radiology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Joan C Vilanova
- Department of Radiology, Clínica Girona, IDI, University of Girona, Girona, Spain
| | - Antonio Luna
- Advanced Medical Imaging, Clinica Las Nieves, SERCOSA (Servicio Radiologia Computerizada), Grupo Health Time, Jaén, Spain; Department of Radiology, Case Western Reserve University, Cleveland, OH
| | - Laura Oleaga
- Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain
| | - Antonio Gómez-Caamaño
- Department of Radiotherapy, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Dow-Mu Koh
- Functional Imaging, Royal Marsden Hospital, Sutton, Surrey, England, UK
| |
Collapse
|
14
|
Terry SYA, Abiraj K, Lok J, Gerrits D, Franssen GM, Oyen WJG, Boerman OC. Can 111In-RGD2 monitor response to therapy in head and neck tumor xenografts? J Nucl Med 2014; 55:1849-55. [PMID: 25349221 DOI: 10.2967/jnumed.114.144394] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
UNLABELLED RGD (arginylglycylaspartic acid)-based imaging tracers allow specific imaging of integrin αvβ3 expression, proteins overexpressed during angiogenesis; however, few studies have investigated the potential of these tracers to monitor responses of antiangiogenic or radiation therapy. In the studies presented here, (111)In-RGD2 was assessed for its potential as an imaging tool to monitor such responses to therapies. METHODS DOTA-E-[c(RGDfK)]2 was radiolabeled with (111)In ((111)In-RGD2), and biodistribution studies were performed in mice with subcutaneous FaDu or SK-RC-52 xenografts after treatment with either antiangiogenic therapy (bevacizumab or sorafenib) or tumor irradiation (10 Gy). Micro-SPECT imaging studies and subsequent quantitative analysis were also performed. The effect of bevacizumab, sorafenib, or radiation therapy on tumor growth was determined. RESULTS The uptake of (111)In-RGD2 in tumors, as determined from biodistribution studies, correlated well with that quantified from micro-SPECT images, and both showed that 15 d after irradiation (111)In-RGD2 uptake was enhanced. Specific or nonspecific uptake of (111)In-RGD2 in FaDu or SK-RC-52 xenografts was not affected after antiangiogenic therapy, except in head and neck squamous cell carcinoma 19 d after the start of sorafenib therapy (P < 0.05). The uptake of (111)In-RGD2 followed tumor volume in studies featuring antiangiogenic therapy. However, the uptake of (111)In-RGD2 in FaDu xenografts was decreased as early as 4 h after tumor irradiation, despite nonspecific uptake remaining unaltered. Tumor growth was inhibited after antiangiogenic or radiation therapy. CONCLUSION Here, it is suggested that (111)In-RGD2 could allow in vivo monitoring of angiogenic responses after radiotherapy and may therefore prove a good clinical tool to monitor angiogenic responses early after the start of radiotherapy in patients with head and neck squamous cell carcinoma. Despite clear antitumor efficacy, antiangiogenic therapy did not alter tumor uptake of (111)In-RGD2, indicating that integrin expression was not altered.
Collapse
Affiliation(s)
- Samantha Y A Terry
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands Department of Imaging Sciences, Kings College London, London, United Kingdom
| | - Keelara Abiraj
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland; and
| | - Jasper Lok
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Danny Gerrits
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben M Franssen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wim J G Oyen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
15
|
Shao Y, Liang W, Kang F, Yang W, Ma X, Li G, Zong S, Chen K, Wang J. A direct comparison of tumor angiogenesis with ⁶⁸Ga-labeled NGR and RGD peptides in HT-1080 tumor xenografts using microPET imaging. Amino Acids 2014; 46:2355-64. [PMID: 24990522 DOI: 10.1007/s00726-014-1788-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 06/13/2014] [Indexed: 11/26/2022]
Abstract
Peptides containing asparagine-glycine-arginine (NGR) and arginine-glycine-aspartic acid (RGD) sequence are being developed for tumor angiogenesis-targeted imaging and therapy. The aim of this study was to compare the efficacy of NGR- and RGD-based probes for imaging tumor angiogenesis in HT-1080 tumor xenografts. Two PET probes, (68)Ga-NOTA-G₃-NGR2 and ⁶⁸Ga-NOTA-G₃-RGD2, were successfully prepared. In vitro stability, partition coefficient, tumor cell binding, as well as in vivo biodistribution properties were also analyzed for both PET probes. The results revealed that the two probes were both hydrophilic and stable in vitro and in vivo, and they were excreted predominately and rapidly through the kidneys. For both probes, the higher tumor uptake and lower accumulation in vital organs were determined. No significant difference between two probes was observed in terms of tumor uptake and the in vivo biodistribution properties. We concluded that these two probes are promising in tumor angiogenesis imaging. ⁶⁸Ga-NOTA-G₃-NGR2 has the potential as an alternative for PET imaging in patients with fibrosarcoma, and it may offer an opportunity to noninvasively monitor CD13-targeted therapy.
Collapse
Affiliation(s)
- Yahui Shao
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 15 Changle West Road, Xi'an, 710032, Shaanxi, China
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Early monitoring antiangiogenesis treatment response of Sunitinib in U87MG Tumor Xenograft by (18)F-FLT MicroPET/CT imaging. BIOMED RESEARCH INTERNATIONAL 2014; 2014:218578. [PMID: 24860813 PMCID: PMC4000939 DOI: 10.1155/2014/218578] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 03/19/2014] [Indexed: 11/17/2022]
Abstract
AIM It was aimed to monitor early treatment response of Sunitinib in U87MG models mimicking glioblastoma multiforme by longitudinal (18)F-FLT microPET/CT imaging in this study. METHODS U87MG tumor mice were intragastrically injected with Sunitinib at a dose of 80 mg/kg for consecutive 7 days. (18)F-FLT microPET/CT scans were acquired on days 0, 1, 3, 7, and 13 after therapy. Tumor sizes and body weight were measured. Tumor samples were collected for immunohistochemical analysis of proliferation and microvessel density (MVD) with anti-Ki67 and anti-CD31, respectively. RESULTS The uptake ratios of tumor to the contralateral muscle (T/M) of (18)F-FLT in the Sunitinib group decreased from baseline to day 3 (T/M0 = 2.98 ± 0.33; T/M3 = 2.23 ± 0.36; P < 0.001), reached the bottom on day 7 (T/M7 = 1.96 ± 0.35; P < 0.001), and then recovered on day 13. The T/M of (18)F-FLT uptake in the control group remained around 3.0. There was no difference for the tumor size between both groups until day 11. (18)F-FLT uptakes of tumor were correlated with Ki67 staining index and MVD. CONCLUSION Early therapy response to Sunitinib could be predicted via (18)F-FLT PET, which will contribute to monitoring antiangiogenesis treatment.
Collapse
|