1
|
Ma X, Mao M, He J, Liang C, Xie HY. Nanoprobe-based molecular imaging for tumor stratification. Chem Soc Rev 2023; 52:6447-6496. [PMID: 37615588 DOI: 10.1039/d3cs00063j] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
The responses of patients to tumor therapies vary due to tumor heterogeneity. Tumor stratification has been attracting increasing attention for accurately distinguishing between responders to treatment and non-responders. Nanoprobes with unique physical and chemical properties have great potential for patient stratification. This review begins by describing the features and design principles of nanoprobes that can visualize specific cell types and biomarkers and release inflammatory factors during or before tumor treatment. Then, we focus on the recent advancements in using nanoprobes to stratify various therapeutic modalities, including chemotherapy, radiotherapy (RT), photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), ferroptosis, and immunotherapy. The main challenges and perspectives of nanoprobes in cancer stratification are also discussed to facilitate probe development and clinical applications.
Collapse
Affiliation(s)
- Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, P. R. China.
| |
Collapse
|
2
|
Design, Synthesis and Cytotoxicity Screening of New Thiazole Derivatives as Potential Anticancer Agents through VEGFR-2 Inhibition. Symmetry (Basel) 2022. [DOI: 10.3390/sym14091814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Z-configurated isomers are kinetically preferred molecules. Compounds with Z-configuration are contained in many natural products, biologically active compounds and as synthons for organic synthesis. Two series of new thiazole-based analogs were synthesized from appropriate starting materials hydrazinecarbothioamide derivatives (Z)-2a,b to be evaluated for their inhibitory activity towards VEGFR-2. The prepared thiazole compounds 3a-5b were screened for their cytotoxic potency against the MDA-MB-231 breast cancer cell line and their percentage inhibition against VEGFR-2. Compound 4d exhibited good VEGFR-2 inhibitory activity. A DNA flow cytometry analysis was conducted, and compound 4d demonstrated cell cycle arrest at the G1 and G2/M phases of the cell cycle profile and an apoptosis-inducing effect by increasing the percentage of pre-G1 phase. Compound 4d was further evaluated for its apoptosis-inducing effect by studying the effect on mitochondrial membrane potential (MMP) and p53 activation. It was found to boost the level of p53 and reduce the level of MMP compared with the untreated control cells.
Collapse
|
3
|
Wang Y, Feng Y, Yang X, Wang W, Wang Y. Diagnosis of Atherosclerotic Plaques Using Vascular Endothelial Growth Factor Receptor-2 Targeting Antibody Nano-microbubble as Ultrasound Contrast Agent. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6524592. [PMID: 35572831 PMCID: PMC9098277 DOI: 10.1155/2022/6524592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023]
Abstract
The atherosclerotic plaque is characterized by narrowing of blood vessels and reduced blood flow leading to the insufficient blood supply to the brain. The hemodynamic changes caused by arterial stenosis increase the shearing force of the fibrous cap on the surface of the plaque, thereby reducing the stability of the plaque. Unstable plaques are more likely to promote angiogenesis and increase the risk of patients with cerebrovascular diseases. A timely understanding of the formation and stability of the arterial plaque can guide in taking targeted measures for reducing the risk of acute stroke in patients. It has been confirmed that nano-microbubbles can enter these plaques through the gaps in the patient's vascular endothelial cells, thereby enhancing the acquisition of ultrasound information for plaque visualization. Therefore, we aim to investigate the diagnostic value of targeted nano-microbubbles for atherosclerotic plaques. This study constructed vascular endothelial growth factor receptor-2 (VEGFR-2) targeting antibody nano-microbubbles and compared its diagnostic value with that of blank nano-microbubbles for atherosclerotic plaques. Studies have found that VEGFR-2 targeting antibody nano-microbubbles can accurately detect the position of plaques. Its detection rate, sensitivity, and specificity for plaques are higher than those of blank nano-microbubbles. Similarly, the peak intensity and average transit time of VEGFR-2 targeting antibody nano-microbubbles were greater than those of blank nano-microbubbles. Therefore, we believe that the combination of VEGFR-2 antibody and nano-microbubbles can enhance the acquisition of ultrasound information on atherosclerotic plaque neovascularization, thereby improving the early diagnosis of unstable plaque.
Collapse
Affiliation(s)
- Yi Wang
- Department of Ultrasonography, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei, China
| | - Yujin Feng
- Department of Ultrasonography, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei, China
| | - Xiaoyun Yang
- Department of Ultrasonography, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei, China
| | - Wengang Wang
- Department of Ultrasonography, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei, China
| | - Yueheng Wang
- Department of Ultrasonography, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei, China
| |
Collapse
|
4
|
Li X, Xia S, Ji R, Zhan W, Zhou W. Evaluation of Microwave Ablation in 4T1 Breast Tumor by a Novel VEFGR2 Targeted Ultrasound Contrast Agents. Front Oncol 2021; 11:690152. [PMID: 34354946 PMCID: PMC8329532 DOI: 10.3389/fonc.2021.690152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/28/2021] [Indexed: 02/01/2023] Open
Abstract
Objectives A novel ultrasound contrast agent (UCA) VEGFR2-targeting iron-doped silica (SiO2) hollow nanoparticles (VEGFR2-PEG-HSNs-Fe NPs) was prepared and applied in microwave ablation for breast cancer to investigate its value in the evaluation of effectiveness after tumor ablation. Methods VEGFR2-PEG-HSNs-Fe NPs were prepared by using nano-SiO2, which was regarded as a substrate and etched by ferrous acetate, and then modified with anti-VEGFR2 antibody. Laser confocal microscope and flow cytometry were used to observe its main physicochemical properties, and biological safety was also investigated. After the xenograft tumor was treated with microwave ablation, the extent of perfusion defect was evaluated by ultrasound by injecting VEGFR2-PEG-HSNs-Fe NPs. Results The average particle size of VEGFR2-PEG-HSNs-Fe was 276.64 ± 30.31 nm, and the surface potential was −13.46 ± 2.83 mV. In vitro, the intensity of ultrasound signal increased with UCA concentration. Good biosafety was performed in in vivo and in vitro experiments. The enhanced ultrasound signal was detected in tumors after injection of VEGFR2-PEG-HSNs-Fe NPs, covering the whole tumor. The lesions, which were incompletely ablated, presented as contrast agent perfusion at the periphery of the tumor, and contrast enhanced ultrasound (CEUS) was performed again after complementary ablation. It was confirmed that all the lesions were completely ablated. Conclusion Nano-targeted UCAs VEGFR2-PEG-HSNs-Fe NPs had good biosafety and ability of specific imaging, which might be used as a contrast agent in CEUS to evaluate the efficacy of tumor ablation.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Ultrasound, RuiJin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shujun Xia
- Department of Ultrasound, RuiJin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ri Ji
- Department of Ultrasound, RuiJin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Weiwei Zhan
- Department of Ultrasound, RuiJin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Zhou
- Department of Ultrasound, RuiJin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Department of Ultrasound, RuiJin Hospital/Lu Wan Branch, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
5
|
Yang HK, Kim JH, Lee HJ, Moon H, Ryu H, Han JK. Early response evaluation of doxorubicin-nanoparticle-microbubble therapy in orthotopic hepatocellular carcinoma rat model using contrast-enhanced ultrasound and intravoxel incoherent motion-diffusion MRI. ULTRASONOGRAPHY (SEOUL, KOREA) 2021; 41:150-163. [PMID: 34304481 PMCID: PMC8696148 DOI: 10.14366/usg.21036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/10/2021] [Indexed: 11/11/2022]
Abstract
Purpose This study aimed to apply doxorubicin-loaded nanoparticle microbubble (Dox-NP-MB) therapy in an orthotopic rat model of hepatocellular carcinoma (HCC) and investigate the utility of contrast-enhanced ultrasound (CEUS) and intravoxel incoherent motion diffusion-weighted magnetic resonance imaging (IVIM-DWI) for response evaluation. Methods Twenty-eight N1S1 HCC model rats were treated with either Dox-NP-MB (group [G] 1, n=8), doxorubicin (Dox) alone (G2, n=7), nanoparticle microbubbles alone (G3, n=7), or saline (G4, control, n=6) on days 0 and 7, and were sacrificed on day 11. IVIM-DWI and CEUS were performed before each treatment and before euthanasia. Efficacy was estimated by the percentage of tumor volume growth inhibition compared with control. Toxicity was assessed by body weight changes and blood tests. Post-treatment changes in IVIM-DWI and CEUS parameters were analyzed. Results Tumor volume growth was inhibited by 48.4% and 90.2% in G1 and G2 compared to G4, respectively. Compared to G2, G1 had a significantly lower degree of body weight change (median, 91.0% [interquartile range, 88.5%-97.0%] vs. 88.0% [82.5%-88.8%], P<0.05) and leukopenia (1.75×103 cells/μL [1.53-2.77] vs. 1.20×103 cells/μL [0.89-1.51], P<0.05). After the first treatment, an increase in peak enhancement, wash-in rate, and wash-in perfusion index on CEUS was observed in G3 and G4 but suppressed in G1 and G2; the apparent diffusion coefficients, true diffusion coefficients, and perfusion fractions significantly increased in G1 and G2 compared to baseline (P<0.05). Conclusion Dox-NP-MB showed reduced Dox toxicity. Early changes in some CEUS and IVIM-DWI parameters correlated with the therapeutic response.
Collapse
Affiliation(s)
- Hyun Kyung Yang
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Radiology, Seoul National University Hospital, Seoul National University, Seoul, Korea
| | - Jung Hoon Kim
- Department of Radiology, Seoul National University Hospital, Seoul National University, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Hak Jong Lee
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Radiology, Seoul National University Bundang Hospital, Seoul National University, Seongnam, Korea
| | | | - Hwaseong Ryu
- Department of Radiology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Joon Koo Han
- Department of Radiology, Seoul National University Hospital, Seoul National University, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
6
|
Synthesis of sorafenib analogues incorporating a 1,2,3-triazole ring and cytotoxicity towards hepatocellular carcinoma cell lines. Bioorg Chem 2021; 112:104831. [PMID: 33831675 DOI: 10.1016/j.bioorg.2021.104831] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/28/2021] [Accepted: 03/12/2021] [Indexed: 01/07/2023]
Abstract
A series of 1,2,3-triazole-containing Sorafenib analogues, in which the aryl urea moiety of Sorafenib (1) was replaced with a 1,2,3-triazole ring linking a substituted phenoxy fragment, were prepared successfully via Huisgen 1,3-dipolar cycloaddition and nucleophilic aromatic substitution. The studies of cytotoxicity towards human hepatocellular carcinoma (HCC) cell lines, HepG2 and Huh7, indicated that p-tert-butylphenoxy analogue 2m showed significant inhibitory activity against Huh7 with IC50 = 5.67 ± 0.57 µM. More importantly, 2m showed low cytotoxicity against human embryonal lung fibroblast cell line, MRC-5, with IC50 > 100 µM, suggesting its highly selective cytotoxic activity (SI > 17.6) towards Huh7 which is much superior to that of Sorafenib (SI = 6.73). The molecular docking studies revealed that the analogue 2m bound B-RAF near the binding position of Sorafenib, while it interacted VEGFR2 efficiently at the same binding position of Sorafenib. However, 2m exhibited moderate inhibitory activity toward B-RAF, implying that its anti-Huh7 effect might not strictly relate to inhibition of B-RAF. Wound healing and BrdU cell proliferation assays confirmed anti-cell migration and anti-cell proliferative activities towards Huh7. With its inhibitory efficiency and high safety profile, 2m has been identified as a promising candidate for the treatment of HCC.
Collapse
|
7
|
Heinen H, Seyler L, Popp V, Hellwig K, Bozec A, Uder M, Ellmann S, Bäuerle T. Morphological, functional, and molecular assessment of breast cancer bone metastases by experimental ultrasound techniques compared with magnetic resonance imaging and histological analysis. Bone 2021; 144:115821. [PMID: 33348127 DOI: 10.1016/j.bone.2020.115821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND The imaging of bone metastases, which is regularly performed by cross-sectional modalities, is clinically vital when characterizing and staging osseous lesions. In this paper, we aimed to establish a novel methodology using experimental ultrasound (US) techniques to assess the morphological, functional, and molecular features of breast cancer bone metastases in an animal model, compared with magnetic resonance imaging (MRI) and histological analysis. MATERIALS AND METHODS Nude rats were implanted intra-arterially with MDA-MB-231 breast cancer cells to induce osteolytic metastasis in their right hind legs. Once tumors had developed, an experimental US technique using automatic 3D scanning and MRI were performed. For assessment of perfusion, functional imaging techniques included contrast-enhanced US (CEUS) and dynamic contrast-enhanced MRI (DCE-MRI). For molecular ultrasound, anti-VEGFR2 conjugated microbubbles were applied and correlated with immunostaining for VEGFR2 expression. RESULTS 3D US enabled the automatic assessment of osteolytic lesions, including the largest tumor diameters along the x-, y- and z-axes as well as the segmented tumor volumes, without significant differences between US and MRI (p > 0.18). The CEUS and DCE-MRI of osseous lesions showed corresponding results for the parameters peak enhancement, wash-in area under the curve (both, r > 0.5) and wash-in perfusion index (r > 0.3) when differentiating between tumor, necrotic tissue and healthy muscle tissue (all, p < 0.01). Finally, molecular US allowed the non-invasive assessment of increased VEGFR2 expression in skeletal lesions compared with surrounding muscle tissue (p = 0.03), while a control antibody could not discriminate between these tissues (p = 0.44)-a factor which was confirmed by histological analysis. CONCLUSION To the best of our knowledge, this is the first report on an imaging protocol for breast cancer bone metastasis using an experimental US scanner. Therefore, we present a novel methodology to characterize these osseous lesions on the morphological, functional, and molecular level in correlation with MRI and histological analysis.
Collapse
Affiliation(s)
- Henrik Heinen
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany; Institute of Radiology, University Hospital, Paracelsus University, Prof.-Ernst-Nathan-Str. 1, 90419 Nuremberg, Germany
| | - Lisa Seyler
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Vanessa Popp
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Konstantin Hellwig
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Aline Bozec
- Medical Clinic 3 - Rheumatology and Immunology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
| | - Michael Uder
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Stephan Ellmann
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany.
| |
Collapse
|
8
|
Bitterer F, Hornung M, Platz Batista da Silva N, Schlitt HJ, Stroszczynski C, Wege AK, Jung EM. In vivo detection of breast cancer liver metastases in humanized tumour mice using tumour specific contrast agent BR55®. Clin Hemorheol Microcirc 2021; 76:559-572. [PMID: 32924994 DOI: 10.3233/ch-200898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To investigate the diagnostic accuracy of high-resolution ultrasound (HRU) for the detection of hepatic metastases of breast cancer in a humanized tumour mouse (HTM) using clinical standard technology. In addition, the efficiency of standard contrast-enhanced ultrasound (CEUS) [microbubbles of sulphur hexafluoride] and CEUS using a novel VEGFR2-targeted contrast agent [BR55®] was examined. METHODS A total of 14 HTM were sonographically examined twice. In addition to a human immune system, the animals developed hepatic tumour lesions after intrahepatic injection of BT-474 breast cancer cells. Digital cine loops from the arterial phase (15-35 sec), the portal venous phase (35-90 sec) and the late phase (3-15 min) of the entire liver were analysed. Data were correlated to histopathology. RESULTS After 9 months, half of the mice (7/14) revealed the development of hepatic breast cancer metastases. The detection limit was 1 mm tumour diameter. In particular, the use of targeted contrast media reduced the needed tumour diameter and helped to precisely classify tumour tissue. In 93% (13/14), the findings of ultrasound could be approved by histological examination by the pathologist. CONCLUSIONS This study in HTM demonstrated the high feasibility of tumour specific contrast media and standard HRU contrast agents to detect early liver metastases.
Collapse
Affiliation(s)
- Florian Bitterer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Hornung
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | | | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | | | - Anja K Wege
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
| | - Ernst-Michael Jung
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
9
|
Lu SL, Liu WW, Cheng JCH, Lin LC, Wang CRC, Li PC. Enhanced Radiosensitization for Cancer Treatment with Gold Nanoparticles through Sonoporation. Int J Mol Sci 2020; 21:ijms21218370. [PMID: 33171604 PMCID: PMC7664670 DOI: 10.3390/ijms21218370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
We demonstrate the megavoltage (MV) radiosensitization of a human liver cancer line by combining gold-nanoparticle-encapsulated microbubbles (AuMBs) with ultrasound. Microbubbles-mediated sonoporation was administered for 5 min, at 2 h prior to applying radiotherapy. The intracellular concentration of gold nanoparticles (AuNPs) increased with the inertial cavitation of AuMBs in a dose-dependent manner. A higher inertial cavitation dose was also associated with more DNA damage, higher levels of apoptosis markers, and inferior cell surviving fractions after MV X-ray irradiation. The dose-modifying ratio in a clonogenic assay was 1.56 ± 0.45 for a 10% surviving fraction. In a xenograft mouse model, combining vascular endothelial growth factor receptor 2 (VEGFR2)-targeted AuMBs with sonoporation significantly delayed tumor regrowth. A strategy involving the spatially and temporally controlled release of AuNPs followed by clinically utilized MV irradiation shows promising results that make it worthy of further translational investigations.
Collapse
Affiliation(s)
- Shao-Lun Lu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan; (S.-L.L.); (W.-W.L.); (J.C.-H.C.); (L.-C.L.)
- Division of Radiation Oncology, National Taiwan University Hospital, Taipei 100229, Taiwan
| | - Wei-Wen Liu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan; (S.-L.L.); (W.-W.L.); (J.C.-H.C.); (L.-C.L.)
| | - Jason Chia-Hsien Cheng
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan; (S.-L.L.); (W.-W.L.); (J.C.-H.C.); (L.-C.L.)
- Division of Radiation Oncology, National Taiwan University Hospital, Taipei 100229, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100229, Taiwan
| | - Lien-Chieh Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan; (S.-L.L.); (W.-W.L.); (J.C.-H.C.); (L.-C.L.)
| | - Churng-Ren Chris Wang
- Department of Chemistry and Biochemistry, National Chung-Cheng University, Chia-Yi 621301, Taiwan;
| | - Pai-Chi Li
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan; (S.-L.L.); (W.-W.L.); (J.C.-H.C.); (L.-C.L.)
- Department of Electrical Engineering, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: ; Tel.: +886-2-3366-3551
| |
Collapse
|
10
|
Antitumor effect of VEGFR2-targeted microbubble destruction with gemcitabine using an endoscopic ultrasound probe: In vivo mouse pancreatic ductal adenocarcinoma model. Hepatobiliary Pancreat Dis Int 2020; 19:478-485. [PMID: 32265136 DOI: 10.1016/j.hbpd.2020.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ultrasound-targeted microbubble destruction (UTMD) induces cellular inflow of drugs at low intensity, while high intensity eradicates tumor vessels. Since vascular endothelial growth factor receptor 2 (VEGFR2) is highly expressed in pancreatic ductal adenocarcinoma (PDAC), VEGFR2-targeted microbubble (MB) might additionally increase the tissue specificity of drugs and thus improve antitumor effects. In addition, fixing the dual pulse intensity could maximize MB properties. This study evaluated the one-off (experiment 1) and cumulative (experiment 2) treatment effect of UTMD by regulating the dual pulse output applied to PDAC using VEGFR2-targeted MB. METHODS C57BL/6 mice inoculated with Pan-02 cells were allocated to five groups: VEGFR2-targeted MB+ gemcitabine (GEM), VEGFR2-targeted MB, non-targeted MB+GEM, GEM, and control groups. After injection of GEM or GEM and either VEGFR2-targeted or non-targeted MB, UTMD was applied for several minutes at low intensity followed by high intensity application. In experiment 1, mice were treated by the protocol described above and then euthanized immediately or at the tumor diameter doubling time (TDT). In experiment 2, the same protocol was repeated weekly and mice were euthanized at TDT regardless of protocol completion. Histological analysis by CD31 and VEGFR2 staining provided microvascular density (MVD) and VEGFR2 expression along vessels (VEGFR2v) or intra/peripheral cells (VEGFR2c). RESULTS In experiment 1, TDT was significantly longer in the VEGFR2-targeted MB+GEM group compared to the non-targeted MB+GEM, GEM, and control groups, while the VEGFR2-targeted MB group showed no statistical significance. MVD and VEGFR2v in the immediate euthanasia was significantly lower in the VEGFR2-targeted MB+GEM and VEGFR2-targeted MB groups than other conditions. In experiment 2, the VEGFR2-targeted MB+GEM group produced significantly longer TDT than the GEM or control groups, whereas the VEGFR2-targeted MB group showed no significant difference. Histology revealed significantly reduced VEGFR2v and VEGFR2c in the VEGFR2-targeted and non-targeted MB+GEM groups, while only VEGFR2v was significantly less in the VEGFR2-targeted MB group. CONCLUSIONS UTMD-mediated GEM therapy with the dual pulse application using VEGFR2-targeted MB substantially suppresses PDCA growth.
Collapse
|
11
|
Piscaglia F, Marcelli E. The expanding potential of functional liver imaging: From research tools to clinical practice in oncology and internal medicine. Eur J Intern Med 2020; 79:23-24. [PMID: 32680652 DOI: 10.1016/j.ejim.2020.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Fabio Piscaglia
- Unit of Internal Medicine, Azienda Ospedaliero Universitaria S.Orsola Malpighi di Bologna, Bologna, Italy.
| | - Emanuela Marcelli
- Laboratory of Bioengineering, Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Turco S, El Kaffas A, Zhou J, Lutz AM, Wijkstra H, Willmann JK, Mischi M. Pharmacokinetic Modeling of Targeted Ultrasound Contrast Agents for Quantitative Assessment of Anti-Angiogenic Therapy: a Longitudinal Case-Control Study in Colon Cancer. Mol Imaging Biol 2020; 21:633-643. [PMID: 30225758 PMCID: PMC6616210 DOI: 10.1007/s11307-018-1274-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE To evaluate quantitative and semi-quantitative ultrasound molecular imaging (USMI) for antiangiogenic therapy monitoring in human colon cancer xenografts in mice. PROCEDURES Colon cancer was established in 17 mice by injection of LS174T (Nr = 9) or CT26 (Nn = 8) cancer cells to simulate clinical responders and non-responders, respectively. Antiangiogenic treatment (bevacizumab; Nrt = Nnt = 5) or control treatment (saline; Nrc = 4, Nnc = 3) was administered at days 0, 3, and 7. Three-dimensional USMI was performed by injection at days 0, 1, 3, 7, and 10 of microbubbles targeted to the vascular endothelial growth factor receptor 2 (VEGFR2). Microbubble binding rate (kb), estimated by first-pass binding model fitting, and semi-quantitative parameters late enhancement (LE) and differential targeted enhancement (dTE) were compared at each day to evaluate their ability to assess and predict the response to therapy. Correlation analysis with the ex-vivo immunohistological quantification of VEGFR2 expression and the percentage blood vessel area was also performed. RESULTS Significant changes in the USMI parameters during treatment were observed only in the responders treated with bevacizumab (p-value < 0.05). Prediction of the response to therapy as early as 1 day after treatment was achieved by the quantitative parameter kb (p-value < 0.01), earlier than possible by tumor volume quantification. USMI parameters could significantly distinguish between clinical responders and non-responders (p-value << 0.01) and correlated well with the ex-vivo quantification of VEGFR2 expression and the percentage blood vessels area (p-value << 0.01). CONCLUSION USMI (semi)quantitative parameters provide earlier assessment of the response to therapy compared to tumor volume, permit early prediction of non-responders, and correlate well with ex-vivo angiogenesis biomarkers.
Collapse
Affiliation(s)
- Simona Turco
- Department of Electrical Engineering, Eindhoven University of Technology, Groene Loper 19, 5612 AZ, Eindhoven, The Netherlands.
| | - Ahmed El Kaffas
- Department of Radiology, Stanford Medicine, Stanford, CA, 94305, USA
| | - Jianhua Zhou
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Amelie M Lutz
- Department of Radiology, Stanford Medicine, Stanford, CA, 94305, USA
| | - Hessel Wijkstra
- Department of Electrical Engineering, Eindhoven University of Technology, Groene Loper 19, 5612 AZ, Eindhoven, The Netherlands
- Department of Urology, Academic Medical Center, 1105 AZ, Amsterdam, The Netherlands
| | - Jürgen K Willmann
- Department of Radiology, Stanford Medicine, Stanford, CA, 94305, USA
| | - Massimo Mischi
- Department of Electrical Engineering, Eindhoven University of Technology, Groene Loper 19, 5612 AZ, Eindhoven, The Netherlands
| |
Collapse
|
13
|
Ultrasound Molecular Imaging of Renal Cell Carcinoma: VEGFR targeted therapy monitored with VEGFR1 and FSHR targeted microbubbles. Sci Rep 2020; 10:7308. [PMID: 32355171 PMCID: PMC7193565 DOI: 10.1038/s41598-020-64433-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/13/2020] [Indexed: 12/15/2022] Open
Abstract
Recent treatment developments for metastatic renal cell carcinoma offer combinations of immunotherapies or immunotherapy associated with tyrosine kinase inhibitors (TKI). There is currently no argument to choose one solution or another. Easy-to-use markers to assess longitudinal responses to TKI are necessary to determine when to switch to immunotherapies. These new markers will enable an earlier adaptation of therapeutic strategy in order to prevent tumor development, unnecessary toxicity and financial costs. This study evaluates the potential of ultrasound molecular imaging to track the response to sunitinib in a clear cell renal carcinoma model (ccRCC). We used a patient-derived xenograft model for this imaging study. Mice harboring human ccRCC were randomized for sunitinib treatment vs. control. The tumors were imaged at days 0, 7, 14 and 28 with ultrasound molecular imaging. Signal enhancement was quantified and compared between the two groups after injections of non-targeted microbubbles and microbubbles targeting VEGFR1 and FSHR. The tumor growth of the sunitinib group was significantly slower. There was a significantly lower expression of both VEGFR-1 and FSHR molecular ultrasound imaging signals in the sunitinib group at all times of treatment (Days 7, 14 and 28). These results confirm the study hypothesis. There was no significant difference between the 2 groups for the non-targeted microbubble ultrasound signal. This study demonstrated for the first time the potential of VEGFR1 and FSHR, by ultrasound-based molecular imaging, to follow-up the longitudinal response to sunitinib in ccRCC. These results should trigger developments for clinical applications.
Collapse
|
14
|
Ultrasound Molecular Imaging With BR55, a Predictive Tool of Antiangiogenic Treatment Efficacy in a Chemo-Induced Mammary Tumor Model. Invest Radiol 2020; 55:657-665. [DOI: 10.1097/rli.0000000000000661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
15
|
Xu ZT, Ding H, Fu TT, Zhu YL, Wang WP. A Nude Mouse Model of Orthotopic Liver Transplantation of Human Hepatocellular Carcinoma HCCLM3 Cell Xenografts and the Use of Imaging to Evaluate Tumor Progression. Med Sci Monit 2019; 25:8694-8703. [PMID: 31736477 PMCID: PMC6880650 DOI: 10.12659/msm.917648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background This study aimed to develop a nude mouse model of orthotopic liver transplantation of HCCLM3 human hepatocellular carcinoma (HCC) cell xenografts and the use of imaging and histology to evaluate tumor development and progression. Material/Methods HCCLM3 cells were injected subcutaneously into 25 healthy male athymic BALB/c (nu/nu) nude mice. The tumors that developed were transplanted into the liver of a new set of nude mice. After four weeks and six weeks, the mice were imaged using ultrasound (US), software-assisted contrast-enhanced ultrasound (CEUS), fluorodeoxyglucose-positron emission tomography (FDG-PET). Histology was performed on the liver and liver tumors, and included immunohistochemistry for vascular endothelial growth factor (VEGF), CD31, CD34, and α-smooth muscle actin (α-SMA). Results The success rate for orthotopic tumor transplantation in the mouse liver was 90% (18/20). Liver tumors measured 11.8±2.6 mm in diameter and 525.9±250.8 mm3 in volume on the sixth week. CEUS showed rapid wash-in and washout in the liver tumors, and PET showed low tumor cell metabolism. Bone metastases were present in 45% (9/20) of mice in the sixth week. Immunohistochemistry showed positive expression for VEGF, CD31, CD34, and α-SMA. Conclusions The nude mouse orthotopic liver transplantation model of human HCC was shown to be a reliable model that has the potential for future research on the pathogenesis and progression of HCC and studies on drug development.
Collapse
Affiliation(s)
- Zhi-Ting Xu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland).,Shanghai Institute of Medical Imaging, Shanghai, China (mainland)
| | - Hong Ding
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland)
| | - Tian-Tian Fu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland).,Shanghai Institute of Medical Imaging, Shanghai, China (mainland)
| | - Yu-Li Zhu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland)
| | - Wen-Ping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
16
|
Rojas JD, Dayton PA. In Vivo Molecular Imaging Using Low-Boiling-Point Phase-Change Contrast Agents: A Proof of Concept Study. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:177-191. [PMID: 30318123 DOI: 10.1016/j.ultrasmedbio.2018.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/26/2018] [Accepted: 08/10/2018] [Indexed: 06/08/2023]
Abstract
Sub-micron phase-change contrast agents (PCCAs) have been proposed as a tool for ultrasound molecular imaging based on their potential to extravasate and target extravascular markers and also because of the potential to image these contrast agents with a high contrast-to-tissue ratio. We compare in vivo ultrasound molecular imaging with targeted low-boiling-point PCCAs and targeted microbubble contrast agents. Both agents were targeted to the intravascular (endothelial) integrin αvß3via a cyclic RGD peptide (cyclo-Arg-Gly-Asp-D-Tyr-Cys) mechanism and imaged in vivo in a rodent fibrosarcoma model, which exhibits angiogenic microvasculature. Signal intensity was measured using two different techniques, conventional contrast-specific imaging (amplitude/phase modulation) and a droplet vaporization imaging sequence, which detects the unique signature of vaporizing PCCAs. Data indicate that PCCA-specific imaging is more sensitive to small numbers of bound agents than conventional contrast imaging. However, data also revealed that contrast from targeted microbubbles was greater than that provided by PCCAs. Both control and targeted PCCAs were observed to be retained in tissue post-vaporization, which was expected for targeted agents but not expected for control agents. The exact mechanism underlying this observation remains unknown.
Collapse
Affiliation(s)
- Juan D Rojas
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, North Carolina, USA.
| |
Collapse
|
17
|
Vishal TMD, Ji-Bin LMD, John EP. Applications in Molecular Ultrasound Imaging: Present and Future. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2019. [DOI: 10.37015/audt.2019.190812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
18
|
Zhu L, Wang L, Liu Y, Xu D, Fang K, Guo Y. CAIX aptamer-functionalized targeted nanobubbles for ultrasound molecular imaging of various tumors. Int J Nanomedicine 2018; 13:6481-6495. [PMID: 30410333 PMCID: PMC6199208 DOI: 10.2147/ijn.s176287] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Targeted nanobubbles can penetrate the tumor vasculature and achieve ultrasound molecular imaging (USMI) of tumor parenchymal cells. However, most targeted nanobubbles only achieve USMI of tumor parenchymal cells from one organ, and their distribution, loading ability, and binding ability in tumors are not clear. Therefore, targeted nanobubbles loaded with carbonic anhydrase IX (CAIX) aptamer were fabricated for USMI of various tumors, and the morphological basis of USMI with targeted nanobubbles was investigated. Materials and methods The specificity of CAIX aptamer at the cellular level was measured by immunofluorescence and flow cytometry. Targeted nanobubbles loaded with CAIX aptamer were prepared by a maleimidethiol coupling reaction, and their binding ability to CAIX-positive tumor cells was analyzed in vitro. USMI of targeted and non-targeted nanobubbles was performed in tumor-bearing nude mice. The distribution, loading ability, and binding ability of targeted nanobubbles in xenograft tumor tissues were demonstrated by immunofluorescence. Results CAIX aptamer could specifically bind to CAIX-positive 786-O and Hela cells, rather than CAIX-negative BxPC-3 cells. Targeted nanobubbles loaded with CAIX aptamer had the advantages of small size, uniform distribution, regular shape, and high safety, and they could specifically accumulate around 786-O and Hela cells, while not binding to BxPC-3 cells in vitro. Targeted nanobubbles had significantly higher peak intensity and larger area under the curve than non-targeted nanobubbles in 786-O and Hela xenograft tumor tissues, while there was no significant difference in the imaging effects of targeted and non-targeted nanobubbles in BxPC-3 xenograft tumor tissues. Immunofluorescence demonstrated targeted nanobubbles could still load CAIX aptamer after penetrating the tumor vasculature and specifically binding to CAIX-positive tumor cells in xenograft tumor tissues. Conclusion Targeted nanobubbles loaded with CAIX aptamer have a good imaging effect in USMI of tumor parenchymal cells, and can improve the accuracy of early diagnosis of malignant tumors from various organs.
Collapse
Affiliation(s)
- Lianhua Zhu
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Shapingba District, Chongqing, China,
| | - Luofu Wang
- Department of Urology, Daping Hospital, Third Military Medical University (Army Medical University), Yuzhong District, Chongqing, China
| | - Yu Liu
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Shapingba District, Chongqing, China,
| | - Dan Xu
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Shapingba District, Chongqing, China,
| | - Kejing Fang
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Shapingba District, Chongqing, China,
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Shapingba District, Chongqing, China,
| |
Collapse
|
19
|
Wischhusen J, Wilson KE, Delcros JG, Molina-Peña R, Gibert B, Jiang S, Ngo J, Goldschneider D, Mehlen P, Willmann JK, Padilla F. Ultrasound molecular imaging as a non-invasive companion diagnostic for netrin-1 interference therapy in breast cancer. Theranostics 2018; 8:5126-5142. [PMID: 30429890 PMCID: PMC6217066 DOI: 10.7150/thno.27221] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023] Open
Abstract
In ultrasound molecular imaging (USMI), ligand-functionalized microbubbles (MBs) are used to visualize vascular endothelial targets. Netrin-1 is upregulated in 60% of metastatic breast cancers and promotes tumor progression. A novel netrin-1 interference therapy requires the assessment of netrin-1 expression prior to treatment. In this study, we studied netrin-1 as a target for USMI and its potential as a companion diagnostic in breast cancer models. Methods: To verify netrin-1 expression and localization, an in vivo immuno-localization approach was applied, in which anti-netrin-1 antibody was injected into living mice 24 h before tumor collection, and revealed with secondary fluorescent antibody for immunofluorescence analysis. Netrin-1 interactions with the cell surface were studied by flow cytometry. Netrin-1-targeted MBs were prepared using MicroMarker Target-Ready (VisualSonics), and validated in in vitro binding assays in static conditions or in a flow chamber using purified netrin-1 protein or netrin-1-expressing cancer cells. In vivo USMI of netrin-1 was validated in nude mice bearing human netrin-1-positive SKBR7 tumors or weakly netrin-1-expressing MDA-MB-231 tumors using the Vevo 2100 small animal imaging device (VisualSonics). USMI feasibility was further tested in transgenic murine FVB/N Tg(MMTV/PyMT634Mul) (MMTV-PyMT) mammary tumors. Results: Netrin-1 co-localized with endothelial CD31 in netrin-1-positive breast tumors. Netrin-1 binding to the surface of endothelial HUVEC and cancer cells was partially mediated by heparan sulfate proteoglycans. MBs targeted with humanized monoclonal anti-netrin-1 antibody bound to netrin-1-expressing cancer cells in static and dynamic conditions. USMI signal was significantly increased with anti-netrin-1 MBs in human SKBR7 breast tumors and transgenic murine MMTV-PyMT mammary tumors compared to signals recorded with either isotype control MBs or after blocking of netrin-1 with humanized monoclonal anti-netrin-1 antibody. In weakly netrin-1-expressing human tumors and normal mammary glands, no difference in imaging signal was observed with anti-netrin-1- and isotype control MBs. Ex vivo analysis confirmed netrin-1 expression in MMTV-PyMT tumors. Conclusions: These results show that USMI allowed reliable detection of netrin-1 on the endothelium of netrin-1-positive human and murine tumors. Significant differences in USMI signal for netrin-1 reflected the significant differences in netrin-1 mRNA & protein expression observed between different breast tumor models. The imaging approach was non-invasive and safe, and provided the netrin-1 expression status in near real-time. Thus, USMI of netrin-1 has the potential to become a companion diagnostic for the stratification of patients for netrin-1 interference therapy in future clinical trials.
Collapse
|
20
|
Shan R, Wang B, Wang A, Sun Z, Dong F, Liu J, Sun H. Endoglin-targeted contrast-enhanced ultrasound imaging in hepatoblastoma xenografts. Oncol Lett 2018; 16:3784-3790. [PMID: 30127989 PMCID: PMC6096263 DOI: 10.3892/ol.2018.9067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 06/04/2018] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is required for the growth of hepatoblastoma (HB). In the present study, an ultrasonic contrast agent, microbubbles (MB), was combined with an endoglin antibody, and then injected into nude mice with HB. This was conducted to detect specific binding to microvessels via non-linear harmonic imaging for tumor angiogenesis assessment. In addition, endoglin expression in experimental animals was measured using western blotting, reverse transcription-quantitative polymerase chain reaction and immunohistochemistry. In vitro, human umbilical vein endothelial cells (HUVECs) were co-cultured with conditioned media collected from HepG2 cells. Western blotting and reverse transcription-quantitative PCR was performed to detect the changes of endoglin expression. In targeted ultrasound imaging, it was determined that the differential targeted enhancement of MBendoglin was significantly higher than that of MBisotype. Over expression of endoglin was identified in the tumor of experimental nude mice; however, it was not present in the liver of the mice. Endoglin expression in HUVECs was significantly increased by co-culture with the conditioned media of HepG2 cells; therefore, the results suggest that endoglin is upregulated in angiogenic vessels in the HepG2 cell xenografts in nude mice. Thus, endoglin-targeted ultrasound imaging is presented as a potential approach for the diagnosis of liver carcinoma.
Collapse
Affiliation(s)
- Rong Shan
- Department of Ultrasonography, Jinan Infectious Disease Hospital, Jinan, Shandong 250021, P.R. China.,Department of Ultrasound, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Bei Wang
- Department of Ultrasonography, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Aiguang Wang
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Zongguo Sun
- The Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Fengyun Dong
- The Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Ju Liu
- The Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Hongjun Sun
- Department of Ultrasonography, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
21
|
Czernuszewicz TJ, Papadopoulou V, Rojas JD, Rajamahendiran RM, Perdomo J, Butler J, Harlacher M, O’Connell G, Zukić D, Aylward SR, Dayton PA, Gessner RC. A new preclinical ultrasound platform for widefield 3D imaging of rodents. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2018; 89:075107. [PMID: 30068108 PMCID: PMC6045495 DOI: 10.1063/1.5026430] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Noninvasive in vivo imaging technologies enable researchers and clinicians to detect the presence of disease and longitudinally study its progression. By revealing anatomical, functional, or molecular changes, imaging tools can provide a near real-time assessment of important biological events. At the preclinical research level, imaging plays an important role by allowing disease mechanisms and potential therapies to be evaluated noninvasively. Because functional and molecular changes often precede gross anatomical changes, there has been a significant amount of research exploring the ability of different imaging modalities to track these aspects of various diseases. Herein, we present a novel robotic preclinical contrast-enhanced ultrasound system and demonstrate its use in evaluating tumors in a rodent model. By leveraging recent advances in ultrasound, this system favorably compares with other modalities, as it can perform anatomical, functional, and molecular imaging and is cost-effective, portable, and high throughput, without using ionizing radiation. Furthermore, this system circumvents many of the limitations of conventional preclinical ultrasound systems, including a limited field-of-view, low throughput, and large user variability.
Collapse
Affiliation(s)
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
| | - Juan D. Rojas
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
| | | | - Jonathan Perdomo
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
| | - James Butler
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
| | - Max Harlacher
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
| | - Graeme O’Connell
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
| | - Dženan Zukić
- Kitware, Inc., Carrboro, North Carolina 27510, USA
| | | | - Paul A. Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
| | - Ryan C. Gessner
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
- Author to whom correspondence should be addressed: . Current address: First Flight Venture Center, 2 Davis Dr., Research Triangle Park, NC 27709-3169. Telephone: 844-766-6865 x707
| |
Collapse
|
22
|
Zhang B, Chen M, Zhang Y, Chen W, Zhang L, Chen L. An ultrasonic nanobubble-mediated PNP/fludarabine suicide gene system: A new approach for the treatment of hepatocellular carcinoma. PLoS One 2018; 13:e0196686. [PMID: 29718963 PMCID: PMC5931662 DOI: 10.1371/journal.pone.0196686] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/17/2018] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE The purpose of this study is to generate an ultrasonic nanobubble (NB)-mediated purine nucleoside phosphorylase (PNP)/fludarabine suicide gene system for the treatment of human hepatocellular carcinoma (HCC). METHODS NBs were prepared from a mixture the phospholipids 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and 1,2-dipalmitoyl-sn-glycero-3-phosphate (DPPA), perfluoropropane gas and other materials using the high shear dispersion method. NBs treated with ultrasound irradiation functioned as a gene-transfer system, and a self-constructed suicide gene expression plasmid, pcDNA3.1(+)/PNP, treated with fludarabine functioned as a therapeutic gene. This system was used to determine the cytotoxic effects of PNP/fludarabine on HepG2 cells and SMMC7721 cells. RESULTS 1. NBs with a small diameter (208-416 nm) and at a high concentration and fine homogeneity were prepared under the optimal method. 2. The pcDNA3.1(+)/PNP plasmid was efficiently transfected into HCC cells using ultrasonic NBs. 3. At 0.75μg/ml fludarabine, PNP/fludarabine showed marked cytotoxic effects toward HepG2 and SMMC7721 cells. PNP/fludarabine achieved the same effect against both SMMC7721 and HepG2 cells but at a lower concentration of fludarabine for the latter. 4. Bystander effects: a 10-20% decrease in the cell survival rate was observed when only 5-10% of transfected cells were PNP positive. CONCLUSIONS NBs constitute a non-toxic, stable and effective gene-delivery platform. The PNP/fludarabine suicide gene system inhibited the growth of HCC cells, induced HCC cell apoptosis, and caused a notable bystander effect at a low fludarabine concentration. This study establishes an important new method for miniaturizing microbubbles and improving a new NB-mediated approach for gene therapy of HCC.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Ultrasonic Imaging, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingna Chen
- Department of Ultrasonic Imaging, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youming Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Chen
- Hepatobiliary and Enteric Surgery Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Zhang
- Hepatobiliary and Enteric Surgery Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Lv Chen
- Department of Occupational and Environmental Health, School of Public Health, Central South University, Changsha, Hunan, PR China
- * E-mail:
| |
Collapse
|
23
|
Rojas JD, Lin F, Chiang YC, Chytil A, Chong DC, Bautch VL, Rathmell WK, Dayton PA. Ultrasound Molecular Imaging of VEGFR-2 in Clear-Cell Renal Cell Carcinoma Tracks Disease Response to Antiangiogenic and Notch-Inhibition Therapy. Theranostics 2018; 8:141-155. [PMID: 29290798 PMCID: PMC5743465 DOI: 10.7150/thno.19658] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 09/14/2017] [Indexed: 12/22/2022] Open
Abstract
Metastatic clear-cell renal cell carcinoma (ccRCC) affects thousands of patients worldwide each year. Antiangiogenic therapy has been shown to have beneficial effects initially, but resistance is eventually developed. Therefore, it is important to accurately track the response of cancer to different therapeutics in order to appropriately adjust the therapy to maximize efficacy. Change in tumor volume is the current gold standard for determining efficacy of treatment. However, functional variations can occur much earlier than measurable volume changes. Contrast-enhanced ultrasound (CEUS) is an important tool for assessing tumor progression and response to therapy, since it can monitor functional changes in the physiology. In this study, we demonstrate how ultrasound molecular imaging (USMI) can accurately track the evolution of the disease and molecular response to treatment. Methods A cohort of NSG (NOD/scid/gamma) mice was injected with ccRCC cells and treated with either the VEGF inhibitor SU (Sunitinib malate, Selleckchem, TX, USA) or the Notch pathway inhibitor GSI (Gamma secretase inhibitor, PF-03084014, Pfizer, New York, NY, USA), or started on SU and later switched to GSI (Switch group). The therapies used in the study focus on disrupting angiogenesis and proper vessel development. SU inhibits signaling of vascular endothelial growth factor (VEGF), which is responsible for the sprouting of new vasculature, and GSI inhibits the Notch pathway, which is a key factor in the correct maturation of newly formed vasculature. Microbubble contrast agents targeted to VEGFR-2 (VEGF Receptor) were delivered as a bolus, and the bound agents were imaged in 3D after the free-flowing contrast was cleared from the body. Additionally, the tumors were harvested at the end of the study and stained for CD31. Results The results show that MI can detect changes in VEGFR-2 expression in the group treated with SU within a week of the start of treatment, while differences in volume only become apparent after the mice have been treated for three weeks. Furthermore, USMI can detect response to therapy in 92% of cases after 1 week of treatment, while the detection rate is only 40% for volume measurements. The amount of targeting for the GSI and Control groups was high throughout the duration of the study, while that of the SU and Switch groups remained low. However, the amount of targeting in the Switch group increased to levels similar to those of the Control group after the treatment was switched to GSI. CD31 staining indicates significantly lower levels of patent vasculature for the SU group compared to the Control and GSI groups. Therefore, the results parallel the expected physiological changes in the tumor, since GSI promotes angiogenesis through the VEGF pathway, while SU inhibits it. Conclusion This study demonstrates that MI can track disease progression and assess functional changes in tumors before changes in volume are apparent, and thus, CEUS can be a valuable tool for assessing response to therapy in disease. Future work is required to determine whether levels of VEGFR-2 targeting correlate with eventual survival outcomes.
Collapse
Affiliation(s)
- Juan D Rojas
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina
| | - Fanglue Lin
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina
| | - Yun-Chen Chiang
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, North Carolina
| | - Anna Chytil
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Diana C Chong
- Curriculum in Genetics and Molecular Biology, The University of North Carolina, Chapel Hill, North Carolina
| | - Victoria L Bautch
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, North Carolina
- Curriculum in Genetics and Molecular Biology, The University of North Carolina, Chapel Hill, North Carolina
- Department of Biology, The University of North Carolina, Chapel Hill, North Carolina
| | - W Kimryn Rathmell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
24
|
Chang EH. An Introduction to Contrast-Enhanced Ultrasound for Nephrologists. Nephron Clin Pract 2017; 138:176-185. [PMID: 29131073 DOI: 10.1159/000484635] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 10/27/2017] [Indexed: 12/15/2022] Open
Abstract
Contrast-enhanced ultrasound (CEUS) is an emerging technology with no known nephrotoxicity. CEUS has been utilized in cardiac and abdominal imaging for decades in Asia and Europe and has recently received greater attention in the United States with its approval for characterization of indeterminate liver lesions. Emerging data suggest that CEUS has potential as a diagnostic imaging tool among individuals who have contraindications to CT and MRI. Few nephrologists are aware of CEUS and even fewer are aware of its potential applications among individuals with kidney disease. This review introduces CEUS to the nephrology community and provides a basic overview of CEUS technology. Knowledge of the applications, advantages, and disadvantages of CEUS provides the framework for nephrologists to make informed decisions regarding this emerging imaging test in appropriate circumstances. This review focuses on the use of CEUS for the characterization of indeterminate kidney lesions and summarizes the most recent data, some of which specifically includes patients with chronic kidney disease (CKD). The results demonstrate that CEUS has high sensitivity and moderate specificity for detecting malignancy in indeterminate kidney lesions among individuals with and without CKD. In conclusion, CEUS is an emerging imaging technique that may have clinically useful applications for detecting malignant kidney lesions, specifically in patients with CKD. However, most of the current data come from small, single-center studies, and larger, multicenter studies are needed.
Collapse
|
25
|
Urnauer S, Müller AM, Schug C, Schmohl KA, Tutter M, Schwenk N, Rödl W, Morys S, Ingrisch M, Bertram J, Bartenstein P, Clevert DA, Wagner E, Spitzweg C. EGFR-targeted nonviral NIS gene transfer for bioimaging and therapy of disseminated colon cancer metastases. Oncotarget 2017; 8:92195-92208. [PMID: 29190908 PMCID: PMC5696174 DOI: 10.18632/oncotarget.21028] [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] [Received: 04/26/2017] [Accepted: 08/04/2017] [Indexed: 02/06/2023] Open
Abstract
Liver metastases present a serious problem in the therapy of advanced colorectal cancer (CRC), as more than 20% of patients have distant metastases at the time of diagnosis with less than 5% being cured. Consequently, new therapeutic approaches are of major need together with high-resolution imaging methods that allow highly specific detection of small metastases. The unique combination of reporter and therapy gene function of the sodium iodide symporter (NIS) may represent a promising theranostic strategy for CRC liver metastases allowing non-invasive imaging of functional NIS expression and therapeutic application of 131I. For targeted NIS gene transfer polymers containing linear polyethylenimine (LPEI), polyethylene glycol (PEG) and the epidermal growth factor receptor (EGFR)-specific ligand GE11 were complexed with human NIS DNA (LPEI-PEG-GE11/NIS). Tumor specificity and transduction efficiency were examined in high EGFR-expressing LS174T metastases by non-invasive imaging using 18F-tetrafluoroborate (18F-TFB) as novel NIS PET tracer. Mice that were injected with LPEI-PEG-GE11/NIS 48 h before 18F-TFB application showed high tumoral levels (4.8±0.6% of injected dose) of NIS-mediated radionuclide uptake in comparison to low levels detected in mice that received untargeted control polyplexes. Three cycles of intravenous injection of EGFR-targeted NIS polyplexes followed by therapeutic application of 55.5 MBq 131I resulted in marked delay in metastases spread, which was associated with improved animal survival. In conclusion, these preclinical data confirm the enormous potential of EGFR-targeted synthetic polymers for systemic NIS gene delivery in an advanced multifocal CRC liver metastases model and open the exciting prospect of NIS-mediated radionuclide therapy in metastatic disease.
Collapse
Affiliation(s)
- Sarah Urnauer
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Andrea M Müller
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Christina Schug
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Kathrin A Schmohl
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Mariella Tutter
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Nathalie Schwenk
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Wolfgang Rödl
- Department of Pharmacy, Center of Drug Research, Pharmaceutical Biotechnology, LMU Munich, Munich, Germany
| | - Stephan Morys
- Department of Pharmacy, Center of Drug Research, Pharmaceutical Biotechnology, LMU Munich, Munich, Germany
| | - Michael Ingrisch
- Department of Clinical Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jens Bertram
- Department of Nuclear Medicine, Radiopharmacy, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Dirk-André Clevert
- Department of Clinical Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ernst Wagner
- Department of Pharmacy, Center of Drug Research, Pharmaceutical Biotechnology, LMU Munich, Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany
| |
Collapse
|
26
|
First-in-Human Ultrasound Molecular Imaging With a VEGFR2-Specific Ultrasound Molecular Contrast Agent (BR55) in Prostate Cancer. Invest Radiol 2017; 52:419-427. [DOI: 10.1097/rli.0000000000000362] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
Lee H, Kim H, Han H, Lee M, Lee S, Yoo H, Chang JH, Kim H. Microbubbles used for contrast enhanced ultrasound and theragnosis: a review of principles to applications. Biomed Eng Lett 2017; 7:59-69. [PMID: 30603152 PMCID: PMC6208473 DOI: 10.1007/s13534-017-0016-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 12/26/2016] [Accepted: 01/18/2017] [Indexed: 12/31/2022] Open
Abstract
Ultrasound was developed several decades ago as a useful imaging modality, and it became the second most popular diagnostic tool due to its non-invasiveness, real-time capabilities, and safety. Additionally, ultrasound has been used as a therapeutic tool with several therapeutic agents and in nanomedicine. Ultrasound imaging is often used to diagnose many types of cancers, including breast, stomach, and thyroid cancers. In addition, ultrasound-mediated therapy is used in cases of joint inflammation, rheumatoid arthritis, and osteoarthritis. Microbubbles, when used as ultrasound contrast agents, can act as echo-enhancers and therapeutic agents, and they can play an essential role in ultrasound imaging and ultrasound-mediated therapy. Recently, various types of ultrasound contrast agents made of lipid, polymer, and protein shells have been used. Air, nitrogen, and perfluorocarbon are usually included in the core of the microbubbles to enhance ultrasound imaging, and therapeutic drugs are conjugated and loaded onto the surface or into the core of the microbubbles, depending on the purpose and properties of the substance. Many research groups have utilized ultrasound contrast agents to enhance the imaging signal in blood vessels or tissues and to overcome the blood-brain barrier or blood-retina barrier. These agents are also used to help treat diseases in various regions or systems of the body, such as the cardiovascular system, or as a cancer treatment. In addition, with the introduction of targeted moiety and multiple functional groups, ultrasound contrast agents are expected to have a potential future in ultrasound imaging and therapy. In this paper, we briefly review the principles of ultrasound and introduce the underlying theory, applications, limitations, and future perspectives of ultrasound contrast agents.
Collapse
Affiliation(s)
- Hohyeon Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Haemin Kim
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Hyounkoo Han
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Minji Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Sunho Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Hongkeun Yoo
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Jin Ho Chang
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
- Sogang Institute of Advanced Technology, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| |
Collapse
|
28
|
Turco S, Tardy I, Frinking P, Wijkstra H, Mischi M. Quantitative ultrasound molecular imaging by modeling the binding kinetics of targeted contrast agent. Phys Med Biol 2017; 62:2449-2464. [DOI: 10.1088/1361-6560/aa5e9a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
29
|
Fornari F, Pollutri D, Patrizi C, La Bella T, Marinelli S, Casadei Gardini A, Marisi G, Baron Toaldo M, Baglioni M, Salvatore V, Callegari E, Baldassarre M, Galassi M, Giovannini C, Cescon M, Ravaioli M, Negrini M, Bolondi L, Gramantieri L. In Hepatocellular Carcinoma miR-221 Modulates Sorafenib Resistance through Inhibition of Caspase-3-Mediated Apoptosis. Clin Cancer Res 2017; 23:3953-3965. [PMID: 28096271 DOI: 10.1158/1078-0432.ccr-16-1464] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 11/29/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023]
Abstract
Purpose: The aberrant expression of miR-221 is a hallmark of human cancers, including hepatocellular carcinoma (HCC), and its involvement in drug resistance, together with a proved in vivo efficacy of anti-miR-221 molecules, strengthen its role as an attractive target candidate in the oncologic field. The discovery of biomarkers predicting the response to treatments represents a clinical challenge in the personalized treatment era. This study aimed to investigate the possible role of miR-221 as a circulating biomarker in HCC patients undergoing sorafenib treatment as well as to evaluate its contribution to sorafenib resistance in advanced HCC.Experimental Design: A chemically induced HCC rat model and a xenograft mouse model, together with HCC-derived cell lines were employed to analyze miR-221 modulation by Sorafenib treatment. Data from the functional analysis were validated in tissue samples from surgically resected HCCs. The variation of circulating miR-221 levels in relation to Sorafenib treatment were assayed in the animal models and in two independent cohorts of patients with advanced HCC.Results: MiR-221 over-expression was associated with Sorafenib resistance in two HCC animal models and caspase-3 was identified as its target gene, driving miR-221 anti-apoptotic activity following Sorafenib administration. Lower pre-treatment miR-221 serum levels were found in patients subsequently experiencing response to Sorafenib and an increase of circulating miR-221 at the two months assessment was observed in responder patients.Conclusions: MiR-221 might represent a candidate biomarker of likelihood of response to Sorafenib in HCC patients to be tested in future studies. Caspase-3 modulation by miR-221 participates to Sorafenib resistance. Clin Cancer Res; 23(14); 3953-65. ©2017 AACR.
Collapse
Affiliation(s)
- Francesca Fornari
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy. .,Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy
| | - Daniela Pollutri
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Clarissa Patrizi
- Center for Regenerative Medicine, Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Tiziana La Bella
- INSERM, UMR-1162, Functional Genomics of Solid Tumors, Paris, France
| | - Sara Marinelli
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Andrea Casadei Gardini
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Marco Baron Toaldo
- Department of Veterinary Medical Sciences, Bologna University, Bologna, Italy
| | - Michele Baglioni
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Veronica Salvatore
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Elisa Callegari
- Department of Morphology, Surgery and Experimental Medicine, Ferrara University, Ferrara, Italy
| | - Maurizio Baldassarre
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Marzia Galassi
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Catia Giovannini
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy.,Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy
| | - Matteo Cescon
- Department of Medical and Surgical Sciences, General and Transplant Surgery Unit, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Matteo Ravaioli
- Department of Medical and Surgical Sciences, General and Transplant Surgery Unit, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Massimo Negrini
- Department of Morphology, Surgery and Experimental Medicine, Ferrara University, Ferrara, Italy
| | - Luigi Bolondi
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy.,Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy
| | - Laura Gramantieri
- Center for Applied Biomedical Research, St.Orsola-Malpighi University Hospital, Bologna, Italy.
| |
Collapse
|
30
|
Eschbach RS, Clevert DA, Hirner-Eppeneder H, Ingrisch M, Moser M, Schuster J, Tadros D, Schneider M, Kazmierczak PM, Reiser M, Cyran CC. Contrast-Enhanced Ultrasound with VEGFR2-Targeted Microbubbles for Monitoring Regorafenib Therapy Effects in Experimental Colorectal Adenocarcinomas in Rats with DCE-MRI and Immunohistochemical Validation. PLoS One 2017; 12:e0169323. [PMID: 28060884 PMCID: PMC5217974 DOI: 10.1371/journal.pone.0169323] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES To investigate contrast-enhanced ultrasound (CEUS) with VEGFR2-targeted microbubbles for monitoring therapy effects of regorafenib on experimental colon carcinomas in rats with correlation to dynamic contrast-enhanced MRI (DCE-MRI) and immunohistochemistry. MATERIALS AND METHODS Human colorectal adenocarcinoma xenografts (HT-29) were implanted subcutaneously in n = 21 (n = 11 therapy group; n = 10 control group) female athymic nude rats (Hsd: RH-Foxn1rnu). Animals were imaged at baseline and after a one-week daily treatment with regorafenib or a placebo (10 mg/kg bodyweight), using CEUS with VEGFR2-targeted microbubbles and DCE-MRI. In CEUS tumor perfusion was assessed during an early vascular phase (wash-in area under the curve = WiAUC) and VEGFR2-specific binding during a late molecular phase (signal intensity after 8 (SI8min) and 10 minutes (SI10min)), using a conventional 15L8 linear transducer (transmit frequency 7 MHz, dynamic range 80 dB, depth 25 mm). In DCE-MRI functional parameters plasma flow (PF) and plasma volume (PV) were quantified. For validation purposes, CEUS parameters were correlated with DCE-MRI parameters and immunohistochemical VEGFR2, CD31, Ki-67 and TUNEL stainings. RESULTS CEUS perfusion parameter WiAUC decreased significantly (116,989 ± 77,048 a.u. to 30,076 ± 27,095a.u.; p = 0.005) under therapy with no significant changes (133,932 ± 65,960 a.u. to 84,316 ± 74,144 a.u.; p = 0.093) in the control group. In the therapy group, the amount of bound microbubbles in the late phase was significantly lower in the therapy than in the control group on day 7 (SI8min: 283 ± 191 vs. 802 ± 460 a.u.; p = 0.006); SI10min: 226 ± 149 vs. 645 ± 461 a.u.; p = 0.009). PF and PV decreased significantly (PF: 147 ± 58 mL/100 mL/min to 71 ± 15 mL/100 mL/min; p = 0.003; PV: 13 ± 3% to 9 ± 4%; p = 0.040) in the therapy group. Immunohistochemistry revealed significantly fewer VEGFR2 (7.2 ± 1.8 vs. 17.8 ± 4.6; p < 0.001), CD31 (8.1 ± 3.0 vs. 20.8 ± 5.7; p < 0.001) and Ki-67 (318.7 ± 94.0 vs. 468.0 ± 133.8; p = 0.004) and significantly more TUNEL (672.7 ± 194.0 vs. 357.6 ± 192.0; p = 0.003) positive cells in the therapy group. CEUS parameters showed significant (p < 0.05) correlations to DCE-MRI parameters and immunohistochemistry. CONCLUSIONS CEUS with VEGFR2-targeted microbubbles allowed for monitoring regorafenib functional and molecular therapy effects on experimental colorectal adenocarcinomas with a significant decline of CEUS and DCE-MRI perfusion parameters as well as a significant reduction of specifically bound microbubbles under therapy, consistent with a reduced expression of VEGFR2.
Collapse
Affiliation(s)
- Ralf Stefan Eschbach
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
- * E-mail:
| | - Dirk-Andre Clevert
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Heidrun Hirner-Eppeneder
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Michael Ingrisch
- Josef Lissner Laboratory for Biomedical Imaging, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Matthias Moser
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Jessica Schuster
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Dina Tadros
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Moritz Schneider
- Josef Lissner Laboratory for Biomedical Imaging, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Philipp Maximilian Kazmierczak
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Maximilian Reiser
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Clemens C. Cyran
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| |
Collapse
|
31
|
Terzi E, Salvatore V, Negrini G, Piscaglia F. Ongoing challenges in the diagnosis of hepatocellular carcinoma. Expert Rev Gastroenterol Hepatol 2016; 10:451-63. [PMID: 26603785 DOI: 10.1586/17474124.2016.1124758] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In 2001, the European Association for the Study of the Liver (EASL) endorsed the possibility of achieving a non-invasive diagnosis of Hepatocellular Carcinoma (HCC) for the first time. Since then, various refinements of the criteria and techniques capable of achieving this diagnosis and the role of plasma and tissue oncomarkers have been reported in the literature and have been accepted to different extents in various geographical areas. Such tools can also potentially imply prognostic significance. The present article critically discusses some of the most relevant and debated challenges which have emerged in this field, including the role of contrast-enhanced ultrasound, and of hepatocyte-specific magnetic resonance contrast agents, the pitfall of transient hepatic attenuation differences, the reliability of biopsy and the status of biomarkers.
Collapse
Affiliation(s)
- Eleonora Terzi
- a Division of Internal Medicine, Department of Digestive Disease and Internal Medicine, Sant'Orsola-Malpighi Hospital , University of Bologna , Bologna , Italy
| | - Veronica Salvatore
- a Division of Internal Medicine, Department of Digestive Disease and Internal Medicine, Sant'Orsola-Malpighi Hospital , University of Bologna , Bologna , Italy
| | - Giulia Negrini
- a Division of Internal Medicine, Department of Digestive Disease and Internal Medicine, Sant'Orsola-Malpighi Hospital , University of Bologna , Bologna , Italy
| | - Fabio Piscaglia
- a Division of Internal Medicine, Department of Digestive Disease and Internal Medicine, Sant'Orsola-Malpighi Hospital , University of Bologna , Bologna , Italy
| |
Collapse
|
32
|
Malone CD, Mattrey RF, Fetzer DT. Contrast-Enhanced Ultrasound (CEUS) for the Diagnosis and Management of Hepatocellular Carcinoma: Current Status and Future Trends. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s11901-016-0324-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
33
|
Thaiss W, Kaufmann S, Kloth C, Nikolaou K, Bösmüller H, Horger M. VEGFR-2 expression in HCC, dysplastic and regenerative liver nodules, and correlation with pre-biopsy Dynamic Contrast Enhanced CT. Eur J Radiol 2016; 85:2036-2041. [DOI: 10.1016/j.ejrad.2016.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022]
|
34
|
Zhou J, Wang H, Zhang H, Lutz AM, Tian L, Hristov D, Willmann JK. VEGFR2-Targeted Three-Dimensional Ultrasound Imaging Can Predict Responses to Antiangiogenic Therapy in Preclinical Models of Colon Cancer. Cancer Res 2016; 76:4081-9. [PMID: 27206846 DOI: 10.1158/0008-5472.can-15-3271] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/25/2016] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) imaging capabilities to assess responses to anticancer therapies are needed to minimize sampling errors common to two-dimensional approaches as a result of spatial heterogeneity in tumors. Recently, the feasibility and reproducibility of 3D ultrasound molecular imaging (3D USMI) using contrast agents, which target molecular markers, have greatly improved, due to the development of clinical 3D matrix array transducers. Here we report preclinical proof-of-concept studies showing that 3D USMI of VEGFR2/KDR expression accurately gauges longitudinal treatment responses to antiangiogenesis therapy in responding versus nonresponding mouse models of colon cancer. Tumors in these models exhibited differential patterns of VEGFR2-targeted 3D USMI signals during the course of antiangiogenic treatment with bevacizumab. In responding tumors, the VEGFR2 signal decreased as soon as 24 hours after therapy was started, whereas in nonresponding tumors there was no change in signal at any time point. The early decrease in VEGFR2 signal was highly predictive of treatment outcome at the end of therapy. Our results offer preclinical proof that 3D USMI can predict responses to antiangiogenic therapy, warranting further investigation of its clinical translatability to predicting treatment outcomes in patients. Cancer Res; 76(14); 4081-9. ©2016 AACR.
Collapse
Affiliation(s)
- Jianhua Zhou
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, California. Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Huaijun Wang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, California
| | - Huiping Zhang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, California
| | - Amelie M Lutz
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, California
| | - Lu Tian
- Department of Health, Research & Policy, Stanford University, Stanford, California
| | - Dimitre Hristov
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Jürgen K Willmann
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, California.
| |
Collapse
|
35
|
Salvatore V, Gianstefani A, Negrini G, Allegretti G, Galassi M, Piscaglia F. Imaging Diagnosis of Hepatocellular Carcinoma: Recent Advances of Contrast-Enhanced Ultrasonography with SonoVue®. Liver Cancer 2016; 5:55-66. [PMID: 29234627 PMCID: PMC5704684 DOI: 10.1159/000367748] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Due to the ability to detect the typical contrast-imaging pattern for hepatocellular carcinoma (HCC), that is hyperenhancement in the arterial phase and hypoenhancement in the late phase on a cirrhotic background, contrast-enhanced ultrasonography (CEUS) was included in the American diagnostic algorithm for HCC in 2005. However, its role has been questioned because of the possibility of misdiagnosis of cholangiocarcinoma. The present review aims to describe the advantages and disadvantages of CEUS applications using Sonovue® for HCC. In particular there is focus on the accuracy of CEUS in detecting the typical HCC pattern, the CEUS patterns of intrahepatic cholangiocarcinoma (ICC), the risk of misdiagnosis with HCC, the diagnostic use of CEUS in cases of locoregional and systemic treatments, and the evaluation of response to antiangiogenic treatment using dedicated software.
Collapse
Affiliation(s)
| | | | | | | | | | - Fabio Piscaglia
- *Fabio Piscaglia, MD, PhD, Division of Internal Medicine, Department of Medical and Surgical Science, University of Bologna, S.Orsola-Malpighi Hospital, Via Albertoni 15, IT-40138, Bologna (Italy), Tel. +39 051 214 2568, E-Mail
| |
Collapse
|
36
|
Payen T, Dizeux A, Baldini C, Le Guillou-Buffello D, Lamuraglia M, Comperat E, Lucidarme O, Bridal SL. VEGFR2-Targeted Contrast-Enhanced Ultrasound to Distinguish between Two Anti-Angiogenic Treatments. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:2202-2211. [PMID: 25980323 DOI: 10.1016/j.ultrasmedbio.2015.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/01/2015] [Accepted: 04/21/2015] [Indexed: 06/04/2023]
Abstract
The aim of this study was to evaluate the capacity of BR55, an ultrasound contrast agent specifically targeting vascular endothelial growth factor receptor 2 (VEGFR2), to distinguish the specific anti-VEGFR2 therapy effect of sunitinib from other anti-angiogenic effects of a therapy (imatinib) that does not directly inhibit VEGFR2. Sunitinib, imatinib and placebo were administered daily for 11 d (264 h) to 45 BalbC mice bearing ectopic CT26 murine colorectal carcinomas. During the course of therapy, B-mode ultrasound, contrast-enhanced ultrasound and VEGFR2-targeted contrast-enhanced ultrasound were performed to assess tumor morphology, vascularization and VEGFR2 expression, respectively. The angiogenic effects on these three aspects were characterized using tumor volume, contrast-enhanced area and differential targeted enhancement. Necrosis, microvasculature and expression of VEGFR2 were also determined by histology and immunostaining. B-Mode imaging revealed that tumor growth was significantly decreased in sunitinib-treated mice at day 11 (p < 0.05), whereas imatinib did not affect growth. Functional evaluation revealed that the contrast-enhanced area decreased significantly (p < 0.02) and by similar amounts under both anti-angiogenic treatments by day 8 (192 h): -23% for imatinib and -21% for sunitinib. No significant decrease was observed in the placebo group. Targeted contrast-enhanced imaging revealed lower differential targeted enhancement, that is, lower levels of VEGFR2 expression, in sunitinib-treated mice relative to placebo-treated mice from 24 h (p < 0.05) and relative to both placebo- and imatinib-treated mice from 48 h (p < 0.05). Histologic assessment of tumors after the final imaging indicated that necrotic area was significantly higher for the sunitinib group (21%) than for the placebo (8%, p < 0.001) and imatinib (11%, p < 0.05) groups. VEGFR2-targeted ultrasound was able to sensitively differentiate the anti-VEGFR2 effect from the reduced area of tumor with functional flow produced by both anti-angiogenic agents. BR55 molecular imaging was, thus, able both to detect early therapeutic response to sunitinib in CT26 tumors as soon as 24 h after the beginning of the treatment and to provide early discrimination (48 h) between tumor response during anti-angiogenic therapy targeting VEGFR2 expression and response during anti-angiogenic therapy not directly acting on this receptor.
Collapse
Affiliation(s)
- Thomas Payen
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France
| | - Alexandre Dizeux
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France
| | - Capucine Baldini
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France
| | | | - Michele Lamuraglia
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France; Medical Oncology Department, Hopital Louis-Mourier, AP-HP, Colombes, France
| | - Eva Comperat
- Anatomic Pathology Department, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Olivier Lucidarme
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, AP-HP, Laboratoire d'Imagerie Biomédicale, Paris, France
| | - S Lori Bridal
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France.
| |
Collapse
|
37
|
Xu J, Lu X, Shi GP. Vasa vasorum in atherosclerosis and clinical significance. Int J Mol Sci 2015; 16:11574-608. [PMID: 26006236 PMCID: PMC4463718 DOI: 10.3390/ijms160511574] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/11/2015] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that leads to several acute cardiovascular complications with poor prognosis. For decades, the role of the adventitial vasa vasorum (VV) in the initiation and progression of atherosclerosis has received broad attention. The presence of VV neovascularization precedes the apparent symptoms of clinical atherosclerosis. VV also mediates inflammatory cell infiltration, intimal thickening, intraplaque hemorrhage, and subsequent atherothrombosis that results in stroke or myocardial infarction. Intraplaque neovessels originating from VV can be immature and hence susceptible to leakage, and are thus regarded as the leading cause of intraplaque hemorrhage. Evidence supports VV as a new surrogate target of atherosclerosis evaluation and treatment. This review provides an overview into the relationship between VV and atherosclerosis, including the anatomy and function of VV, the stimuli of VV neovascularization, and the available underlying mechanisms that lead to poor prognosis. We also summarize translational researches on VV imaging modalities and potential therapies that target VV neovascularization or its stimuli.
Collapse
Affiliation(s)
- Junyan Xu
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
| | - Xiaotong Lu
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
| | - Guo-Ping Shi
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Abstract
In view of the trend towards personalized treatment strategies for (cancer) patients, there is an increasing need to noninvasively determine individual patient characteristics. Such information enables physicians to administer to patients accurate therapy with appropriate timing. For the noninvasive visualization of disease-related features, imaging biomarkers are expected to play a crucial role. Next to the chemical development of imaging probes, this requires preclinical studies in animal tumour models. These studies provide proof-of-concept of imaging biomarkers and help determine the pharmacokinetics and target specificity of relevant imaging probes, features that provide the fundamentals for translation to the clinic. In this review we describe biological processes derived from the “hallmarks of cancer” that may serve as imaging biomarkers for diagnostic, prognostic and treatment response monitoring that are currently being studied in the preclinical setting. A number of these biomarkers are also being used for the initial preclinical assessment of new intervention strategies. Uniquely, noninvasive imaging approaches allow longitudinal assessment of changes in biological processes, providing information on the safety, pharmacokinetic profiles and target specificity of new drugs, and on the antitumour effectiveness of therapeutic interventions. Preclinical biomarker imaging can help guide translation to optimize clinical biomarker imaging and personalize (combination) therapies.
Collapse
|
39
|
Sacco R, Mismas V, Romano A, Bertini M, Bertoni M, Federici G, Metrangolo S, Parisi G, Tumino E, Bresci G, Giacomelli L, Marceglia S, Bargellini I. Assessment of clinical and radiological response to sorafenib in hepatocellular carcinoma patients. World J Hepatol 2015; 7:33-39. [PMID: 25624994 PMCID: PMC4295191 DOI: 10.4254/wjh.v7.i1.33] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/10/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Sorafenib is an effective anti-angiogenic treatment for hepatocellular carcinoma (HCC). The assessment of tumor progression in patients treated with sorafenib is crucial to help identify potentially-resistant patients, avoiding unnecessary toxicities. Traditional methods to assess tumor progression are based on variations in tumor size and provide unreliable results in patients treated with sorafenib. New methods to assess tumor progression such as the modified Response Evaluation Criteria in Solid Tumors or European Association for the Study of Liver criteria are based on imaging to measure the vascularization and tumor volume (viable or necrotic). These however fail especially when the tumor response results in irregular development of necrotic tissue. Newer assessment techniques focus on the evaluation of tumor volume, density or perfusion. Perfusion computed tomography and Dynamic Contrast-Enhanced-UltraSound can measure the vascularization of HCC lesions and help predict tumor response to anti-angiogenic therapies. Mean Transit Time is a possible predictive biomarker to measure tumor response. Volumetric techniques are reliable, reproducible and time-efficient and can help measure minimal changes in viable tumor or necrotic tissue, allowing the prompt identification of non-responders. Volume ratio may be a reproducible biomarker for tumor response. Larger trials are needed to confirm the use of these techniques in the prediction of response to sorafenib.
Collapse
|
40
|
Chuang HY, Chang YF, Liu RS, Hwang JJ. Serial low doses of sorafenib enhance therapeutic efficacy of adoptive T cell therapy in a murine model by improving tumor microenvironment. PLoS One 2014; 9:e109992. [PMID: 25333973 PMCID: PMC4198194 DOI: 10.1371/journal.pone.0109992] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/09/2014] [Indexed: 01/05/2023] Open
Abstract
Requirements of large numbers of transferred T cells and various immunosuppressive factors and cells in the tumor microenvironment limit the applications of adoptive T cells therapy (ACT) in clinic. Accumulating evidences show that chemotherapeutic drugs could act as immune supportive instead of immunosuppressive agents when proper dosage is used, and combined with immunotherapy often results in better treatment outcomes than monotherapy. Controversial immunomodulation effects of sorafenib, a multi-kinases inhibitor, at high and low doses have been reported in several types of cancer. However, what is the range of the low-dose sorafenib will influence the host immunity and responses of ACT is still ambiguous. Here we used a well-established E.G7/OT-1 murine model to understand the effects of serial low doses of sorafenib on both tumor microenvironment and transferred CD8+ T cells and the underlying mechanisms. Sorafenib lowered the expressions of immunosuppressive factors, and enhanced functions and migrations of transferred CD8+ T cells through inhibition of STAT3 and other immunosuppressive factors. CD8+ T cells were transduced with granzyme B promoter for driving imaging reporters to visualize the activation and distribution of transferred CD8+ T cells prior to adoptive transfer. Better activations of CD8+ T cells and tumor inhibitions were found in the combinational group compared with CD8+ T cells or sorafenib alone groups. Not only immunosuppressive factors but myeloid derived suppressive cells (MDSCs) and regulatory T cells (Tregs) were decreased in sorafenib-treated group, indicating that augmentation of tumor inhibition and function of CD8+ T cells by serial low doses of sorafenib were via reversing the immunosuppressive microenvironment. These results revealed that the tumor inhibitions of sorafenib not only through eradicating tumor cells but modifying tumor microenvironment, which helps outcomes of ACT significantly.
Collapse
Affiliation(s)
- Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ya-Fang Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ren-Shyan Liu
- National PET/Cyclotron Center and Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jeng-Jong Hwang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|