1
|
She L, Su L, Liu C. Bevacizumab combined with re-irradiation in recurrent glioblastoma. Front Oncol 2022; 12:961014. [PMID: 36046037 PMCID: PMC9423039 DOI: 10.3389/fonc.2022.961014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
Background Glioblastoma is characterized by rich vasculature and abnormal vascular structure and function. Currently, there is no standard treatment for recurrent glioblastoma (rGBM). Bevacizumab (BEV) has established role of inhibiting neovascularization, alleviating hypoxia in the tumor area and activating the immune microenvironment. BEV may exert synergistic effects with re-irradiation (re-RT) to improve the tumor microenvironment for rGBM. Purpose The purpose of this study was to evaluate the safety, tolerability, and efficacy of a combination of BEV and re-RT for rGBM treatment. Methods In this retrospective study, a total of 26 rGBM patients with surgical pathologically confirmed glioblastoma and at least one event of recurrence were enrolled. All patients were treated with re-RT in combination with BEV. BEV was administered until progression or serious adverse events. Results Median follow-up was 21.9 months for all patients, whereas median progression-free survival (PFS) was 8.0 months (95% confidence interval [CI]: 6.5–9.5 months). In addition, the 6-month and 1-year PFS rates were 65.4% and 28.2%, respectively. The median overall survival (OS), 6-month OS rate, and 1-year OS rate were 13.6 months (95% CI: 10.2–17.0 months), 92.3%, and 67.5%, respectively. The patient showed good tolerance during the treatment with no grade > 3 grade side event and radiation necrosis occurrence rate of 0%. Combined treatment of gross total resection (GTR) before re-RT and concurrent temozolomide during re-RT was an independent prognostic factor that affected both OS and PFS in the whole cohort (OS: 0.067, 95% CI: 0.009–0.521, p = 0.010; PFS: 0.238, 95% CI: 0.076–0.744, p = 0.038). Conclusion In this study, re-RT combined with concurrent and maintenance BEV treatment was safe, tolerable, and effective in rGBM patients. Moreover, GTR before re-RT and selective concurrent temozolomide could further improve patient PFS and OS.
Collapse
Affiliation(s)
- Lei She
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Su
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
2
|
Application of Ultrasound Combined with Microbubbles for Cancer Therapy. Int J Mol Sci 2022; 23:ijms23084393. [PMID: 35457210 PMCID: PMC9026557 DOI: 10.3390/ijms23084393] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023] Open
Abstract
At present, cancer is one of the leading causes of death worldwide. Treatment failure remains one of the prime hurdles in cancer treatment due to the metastatic nature of cancer. Techniques have been developed to hinder the growth of tumours or at least to stop the metastasis process. In recent years, ultrasound therapy combined with microbubbles has gained immense success in cancer treatment. Ultrasound-stimulated microbubbles (USMB) combined with other cancer treatments including radiation therapy, chemotherapy or immunotherapy has demonstrated potential improved outcomes in various in vitro and in vivo studies. Studies have shown that low dose radiation administered with USMB can have similar effects as high dose radiation therapy. In addition, the use of USMB in conjunction with radiotherapy or chemotherapy can minimize the toxicity of high dose radiation or chemotherapeutic drugs, respectively. In this review, we discuss the biophysical properties of USMB treatment and its applicability in cancer therapy. In particular, we highlight important preclinical and early clinical findings that demonstrate the antitumour effect combining USMB and other cancer treatment modalities (radiotherapy and chemotherapy). Our review mainly focuses on the tumour vascular effects mediated by USMB and these cancer therapies. We also discuss several current limitations, in addition to ongoing and future efforts for applying USMB in cancer treatment.
Collapse
|
3
|
Didier RA, Biko DM, Hwang M, Unnikrishnan S, Woźniak MM, Yusuf GT, Sridharan A. Emerging contrast-enhanced ultrasound applications in children. Pediatr Radiol 2021; 51:2418-2424. [PMID: 33791840 DOI: 10.1007/s00247-021-05045-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/27/2021] [Accepted: 03/01/2021] [Indexed: 11/25/2022]
Abstract
Ultrasound contrast agent (UCA) use in radiology is expanding beyond traditional applications such as evaluation of liver lesions, vesicoureteral reflux and echocardiography. Among emerging techniques, 3-D and 4-D contrast-enhanced ultrasound (CEUS) imaging have demonstrated potential in enhancing the accuracy of voiding urosonography and are ready for wider clinical adoption. US contrast-based lymphatic imaging has been implemented for guiding needle placement in MR lymphangiography in children. In adults, intraoperative CEUS imaging has improved diagnosis and assisted surgical management in tumor resection, and its translation to pediatric brain tumor surgery is imminent. Because of growing interest in precision medicine, targeted US molecular imaging is a topic of active preclinical research and early stage clinical translation. Finally, an exciting new development in the application of UCA is in the field of localized drug delivery and release, with a particular emphasis on treating aggressive brain tumors. Under the appropriate acoustic settings, UCA can reversibly open the blood-brain barrier, allowing drug delivery into the brain. The aim of this article is to review the emerging CEUS applications and provide evidence regarding the feasibility of these applications for clinical implementation.
Collapse
Affiliation(s)
- Ryne A Didier
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA.
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - David M Biko
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Misun Hwang
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Unnikrishnan
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Magdalena M Woźniak
- Department of Pediatric Radiology, Medical University of Lublin, Lublin, Poland
| | - Gibran T Yusuf
- Department of Radiology, King's College Hospital, Denmark Hill, London, UK
| | - Anush Sridharan
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| |
Collapse
|
4
|
Molecular Ultrasound Imaging. NANOMATERIALS 2020; 10:nano10101935. [PMID: 32998422 PMCID: PMC7601169 DOI: 10.3390/nano10101935] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
In the last decade, molecular ultrasound imaging has been rapidly progressing. It has proven promising to diagnose angiogenesis, inflammation, and thrombosis, and many intravascular targets, such as VEGFR2, integrins, and selectins, have been successfully visualized in vivo. Furthermore, pre-clinical studies demonstrated that molecular ultrasound increased sensitivity and specificity in disease detection, classification, and therapy response monitoring compared to current clinically applied ultrasound technologies. Several techniques were developed to detect target-bound microbubbles comprising sensitive particle acoustic quantification (SPAQ), destruction-replenishment analysis, and dwelling time assessment. Moreover, some groups tried to assess microbubble binding by a change in their echogenicity after target binding. These techniques can be complemented by radiation force ultrasound improving target binding by pushing microbubbles to vessel walls. Two targeted microbubble formulations are already in clinical trials for tumor detection and liver lesion characterization, and further clinical scale targeted microbubbles are prepared for clinical translation. The recent enormous progress in the field of molecular ultrasound imaging is summarized in this review article by introducing the most relevant detection technologies, concepts for targeted nano- and micro-bubbles, as well as their applications to characterize various diseases. Finally, progress in clinical translation is highlighted, and roadblocks are discussed that currently slow the clinical translation.
Collapse
|
5
|
Cui H, Zhang D, Peng F, Kong H, Guo Q, Wu T, Wen X, Zhang L, Tian J. Identifying ultrasound features of positive expression of Ki67 and P53 in breast cancer using radiomics. Asia Pac J Clin Oncol 2020; 17:e176-e184. [PMID: 32779399 DOI: 10.1111/ajco.13397] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE To examine the relationship between ultrasonic findings and positive expression of Ki67 and P53 in breast cancer. MATERIAL AND METHODS Surgical resection specimens of 263 breast cancer lesions were examined. Ultrasound examination and pathological examination were performed on each lesion for retrospective analysis. We applied regression analysis to the ultrasonic features related to the positive expression of Ki67 and P53 and obtained the corresponding models. To analyze diagnostic efficiency, we calculated the area under the curve (AUC). Additionally, we created a heat map to show the results of the cluster analysis. RESULTS Lesions with higher Ki67 expression were associated with posterior acoustic enhancement, absence of an echo halo and a higher Breast Imaging Reporting and Data System (BI-RADS) category. P53-positive cancer were associated with an absence of an echo halo and a higher BI-RADS category. The AUC of the regression models of Ki67 and P53 was 0.78 and 0.71, respectively. CONCLUSIONS Our study revealed that breast cancer ultrasonic findings were closely related to expression of molecular indicators, suggesting that ultrasound can be used to provide useful information to clinicians.
Collapse
Affiliation(s)
- Hao Cui
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Dandan Zhang
- Department of Ultrasound Medicine, Heilongjiang provincial hospital, Heilongjiang, China
| | - Fuhui Peng
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Hanqing Kong
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Qiang Guo
- Department of Ultrasound Medicine, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Tong Wu
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Xin Wen
- Department of Ultrasound Medicine, the Third Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Lei Zhang
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Jiawei Tian
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| |
Collapse
|
6
|
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
|
7
|
Abstract
Contrast-enhanced ultrasound (CEUS) imaging is a valuable tool for preclinical and clinical diagnostics. The most frequently used ultrasound contrast agents are microbubbles. Besides them, novel nano-sized materials are under investigation, which are briefly discussed in this chapter. For molecular CEUS, the ultrasound contrast agents are modified to actively target disease-associated molecular markers with a site-specific ligand. The most common markers for tumor imaging are related to neoangiogenesis, like the vascular endothelial growth factor receptor-2 (VEGFR2) and αvβ3 integrin. In this chapter, applications of molecular ultrasound to longitudinally monitor receptor expression during tumor growth, to detect neovascularization, and to evaluate therapy responses are described. Furthermore, we report on first clinical trials of molecular CEUS with VEGFR2-targeted phospholipid microbubbles showing promising results regarding patient safety and its ability to detect tumors of prostate, breast, and ovary. The chapter closes with an outlook on ultrasound theranostics, where (targeted) ultrasound contrast agents are used to increase the permeability of tumor tissues and to support drug delivery.
Collapse
Affiliation(s)
- Jasmin Baier
- Institute for Experimental Molecular Imaging Organization University Clinics, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging Organization University Clinics, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging Organization University Clinics, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany.
| |
Collapse
|
8
|
Balasundaram G, Ding L, Li X, Attia ABE, Dean-Ben XL, Ho CJH, Chandrasekharan P, Tay HC, Lim HQ, Ong CB, Mason RP, Razansky D, Olivo M. Noninvasive Anatomical and Functional Imaging of Orthotopic Glioblastoma Development and Therapy using Multispectral Optoacoustic Tomography. Transl Oncol 2018; 11:1251-1258. [PMID: 30103155 PMCID: PMC6092474 DOI: 10.1016/j.tranon.2018.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/26/2018] [Accepted: 07/02/2018] [Indexed: 01/13/2023] Open
Abstract
PURPOSE Here we demonstrate the potential of multispectral optoacoustic tomography (MSOT), a new non-invasive structural and functional imaging modality, to track the growth and changes in blood oxygen saturation (sO2) in orthotopic glioblastoma (GBMs) and the surrounding brain tissues upon administration of a vascular disruptive agent (VDA). METHODS Nude mice injected with U87MG tumor cells were longitudinally monitored for the development of orthotopic GBMs up to 15 days and observed for changes in sO2 upon administration of combretastatin A4 phosphate (CA4P, 30 mg/kg), an FDA approved VDA for treating solid tumors. We employed a newly-developed non-negative constrained approach for combined MSOT image reconstruction and unmixing in order to quantitatively map sO2 in whole mouse brains. RESULTS Upon longitudinal monitoring, tumors could be detected in mouse brains using single-wavelength data as early as 6 days post tumor cell inoculation. Fifteen days post-inoculation, tumors had higher sO2 of 63 ± 11% (n = 5, P < .05) against 48 ± 7% in the corresponding contralateral brain, indicating their hyperoxic status. In a different set of animals, 42 days post-inoculation, tumors had lower sO2 of 42 ± 5% against 49 ± 4% (n = 3, P < .05) in the contralateral side, indicating their hypoxic status. Upon CA4P administration, sO2 in 15 days post-inoculation tumors dropped from 61 ± 9% to 36 ± 1% (n = 4, P < .01) within one hour, then reverted to pre CA4P treatment values (63 ± 6%) and remained constant until the last observation time point of 6 hours. CONCLUSION With the help of advanced post processing algorithms, MSOT was capable of monitoring the tumor growth and assessing hemodynamic changes upon administration of VDAs in orthotopic GBMs.
Collapse
Affiliation(s)
- Ghayathri Balasundaram
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios, Singapore 138667
| | - Lu Ding
- Institute for Biological and Medical Imaging, Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Xiuting Li
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios, Singapore 138667
| | - Amalina Binte Ebrahim Attia
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios, Singapore 138667
| | - Xose Luis Dean-Ben
- Institute for Biological and Medical Imaging, Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Chris Jun Hui Ho
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios, Singapore 138667
| | - Prashant Chandrasekharan
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios, Singapore 138667
| | - Hui Chien Tay
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios, Singapore 138667
| | - Hann Qian Lim
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios, Singapore 138667
| | - Chee Bing Ong
- Advanced Molecular Pathology Lab (AMPL), Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos building, Singapore 138673
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel Razansky
- Institute for Biological and Medical Imaging, Technical University of Munich and Helmholtz Center Munich, Munich, Germany.
| | - Malini Olivo
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios, Singapore 138667.
| |
Collapse
|
9
|
Theek B, Opacic T, Lammers T, Kiessling F. Semi-Automated Segmentation of the Tumor Vasculature in Contrast-Enhanced Ultrasound Data. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:1910-1917. [PMID: 29730066 DOI: 10.1016/j.ultrasmedbio.2018.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/12/2018] [Accepted: 03/16/2018] [Indexed: 05/20/2023]
Abstract
The vascular architecture in tumors contains relevant information for tumor classification and evaluation of therapy responses. To develop a reliable and user-independent analysis tool, a foreground detection algorithm was combined with a maximum-intensity projection to obtain a high signal-to-noise image from contrast-enhanced B-mode data sets, enabling vessel segmentation by thresholding. Parameters describing the density of the vascular network, the number of vessels and the number of branches were extracted. The highly angiogenic A431 tumors had a relative blood volume of 49%, a mean pixel distance to the next vessel of 1.8 ± 0.3 px, 51 ± 29 individual vessels and 478 ± 184 branching points, whereas the more mature and heterogeneous vascularized human epithelial ovarian carcinoma (MLS) and A549 tumors had values of 30%, 3.7 ± 2.7 px, 65 ± 12 and 220 ± 159, and 13%, 7.4 ± 2 px, 31 ± 9 and 59 ± 40, respectively. Thus, our semi-automated analysis method enables the extraction of quantitative vascular features that may help to simplify and standardize tumor characterization.
Collapse
Affiliation(s)
- Benjamin Theek
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Tatjana Opacic
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany.
| |
Collapse
|
10
|
Kim H, Chang JH. Multimodal photoacoustic imaging as a tool for sentinel lymph node identification and biopsy guidance. Biomed Eng Lett 2018; 8:183-191. [PMID: 30603202 PMCID: PMC6208518 DOI: 10.1007/s13534-018-0068-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/14/2018] [Accepted: 04/15/2018] [Indexed: 02/06/2023] Open
Abstract
As a minimally invasive method, sentinel lymph node biopsy (SLNB) in conjunction with guidance methods is the standard method to determine cancer metastasis in breast. The desired guidance methods for SLNB should be capable of precise SLN localization for accurate diagnosis of micro-metastases at an early stage of cancer progression and thus facilitate reducing the number of SLN biopsies for minimal surgical complications. For this, high sensitivity to the administered dyes, high spatial and contrast resolutions, deep imaging depth, and real-time imaging capability are pivotal requirements. Currently, various methods have been used for SLNB guidance, each with their own advantages and disadvantages, but no methods meet the requirements. In this review, we discuss the conventional SLNB guidance methods in this perspective. In addition, we focus on the role of the PA imaging modality on real-time SLN identification and biopsy guidance. In particular, PA-based hybrid imaging methods for precise SLN identification and efficient biopsy guidance are introduced, and their unique features, advantages, and disadvantages are discussed.
Collapse
Affiliation(s)
- Haemin Kim
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 South Korea
| | - Jin Ho Chang
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 South Korea
- Department of Electronic Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 South Korea
| |
Collapse
|
11
|
Wang S, Hossack JA, Klibanov AL. Targeting of microbubbles: contrast agents for ultrasound molecular imaging. J Drug Target 2018; 26:420-434. [PMID: 29258335 DOI: 10.1080/1061186x.2017.1419362] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
For contrast ultrasound imaging, the most efficient contrast agents comprise highly compressible gas-filled microbubbles. These micrometer-sized particles are typically filled with low-solubility perfluorocarbon gases, and coated with a thin shell, often a lipid monolayer. These particles circulate in the bloodstream for several minutes; they demonstrate good safety and are already in widespread clinical use as blood pool agents with very low dosage necessary (sub-mg per injection). As ultrasound is an ubiquitous medical imaging modality, with tens of millions of exams conducted annually, its use for molecular/targeted imaging of biomarkers of disease may enable wider implementation of personalised medicine applications, precision medicine, non-invasive quantification of biomarkers, targeted guidance of biopsy and therapy in real time. To achieve this capability, microbubbles are decorated with targeting ligands, possessing specific affinity towards vascular biomarkers of disease, such as tumour neovasculature or areas of inflammation, ischaemia-reperfusion injury or ischaemic memory. Once bound to the target, microbubbles can be selectively visualised to delineate disease location by ultrasound imaging. This review discusses the general design trends and approaches for such molecular ultrasound imaging agents, which are currently at the advanced stages of development, and are evolving towards widespread clinical trials.
Collapse
Affiliation(s)
- Shiying Wang
- a Department of Biomedical Engineering , University of Virginia , Charlottesville , VA , USA
| | - John A Hossack
- a Department of Biomedical Engineering , University of Virginia , Charlottesville , VA , USA
| | - Alexander L Klibanov
- a Department of Biomedical Engineering , University of Virginia , Charlottesville , VA , USA.,b Cardiovascular Division (Department of Medicine), Robert M Berne Cardiovascular Research Center , University of Virginia , Charlottesville , VA , USA
| |
Collapse
|
12
|
Lux J, Vezeridis AM, Hoyt K, Adams SR, Armstrong AM, Sirsi SR, Mattrey RF. Thrombin-Activatable Microbubbles as Potential Ultrasound Contrast Agents for the Detection of Acute Thrombosis. ACS APPLIED MATERIALS & INTERFACES 2017; 9:37587-37596. [PMID: 28994575 PMCID: PMC5691601 DOI: 10.1021/acsami.7b10592] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Acute deep vein thrombosis (DVT) is the formation of a blood clot in the deep veins of the body that can lead to fatal pulmonary embolism. Acute DVT is difficult to distinguish from chronic DVT by ultrasound (US), the imaging modality of choice, and is therefore treated aggressively with anticoagulants, which can lead to internal bleeding. Here we demonstrate that conjugating perfluorobutane-filled (PFB-filled) microbubbles (MBs) with thrombin-sensitive activatable cell-penetrating peptides (ACPPs) could lead to the development of contrast agents that detect acute thrombosis with US imaging. Successful conjugation of ACPP to PFB-filled MBs was confirmed by fluorescence microscopy and flow cytometry. Fluorescein-labeled ACPP was used to evaluate the efficiency of thrombin-triggered cleavage by measuring the mean fluorescence intensity of ACPP-labeled MBs (ACPP-MBs) before and after incubation at 37 °C with thrombin. Lastly, control MBs and ACPP-MBs were infused through a tube containing a clot, and US contrast enhancement was measured with or without the presence of a thrombin inhibitor after washing the clot with saline. With thrombin activity, 91.7 ± 14.2% of the signal was retained after ACPP-MB infusion and washing, whereas only 16.7 ± 4% of the signal was retained when infusing ACPP-MBs in the presence of hirudin, a potent thrombin inhibitor.
Collapse
Affiliation(s)
- Jacques Lux
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| | - Alexander M. Vezeridis
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| | - Kenneth Hoyt
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Stephen R. Adams
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093, United States
| | - Amanda M. Armstrong
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| | - Shashank R. Sirsi
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Robert F. Mattrey
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| |
Collapse
|
13
|
Chang EL, Ting CY, Hsu PH, Lin YC, Liao EC, Huang CY, Chang YC, Chan HL, Chiang CS, Liu HL, Wei KC, Fan CH, Yeh CK. Angiogenesis-targeting microbubbles combined with ultrasound-mediated gene therapy in brain tumors. J Control Release 2017; 255:164-175. [DOI: 10.1016/j.jconrel.2017.04.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 02/05/2017] [Accepted: 04/09/2017] [Indexed: 01/21/2023]
|
14
|
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
|
15
|
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]
|
16
|
Song JL, Chen C, Yuan JP, Sun SR. Progress in the clinical detection of heterogeneity in breast cancer. Cancer Med 2016; 5:3475-3488. [PMID: 27774765 PMCID: PMC5224851 DOI: 10.1002/cam4.943] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is currently the most common form of cancer and the second‐leading cause of death from cancer in women. Though considerable progress has been made in the treatment of breast cancer, the heterogeneity of tumors (both inter‐ and intratumor) remains a considerable diagnostic and prognostic challenge. From clinical observation to genetic mutations, the history of understanding the heterogeneity of breast cancer is lengthy and detailed. Effectively detecting heterogeneity in breast cancer is important during treatment. Various methods of depicting this heterogeneity are now available and include genetic, pathologic, and imaging analysis. These methods allow characterization of the heterogeneity of breast cancer on a genetic level, providing greater insight during the process of establishing an effective therapeutic plan. This study reviews how the understanding of tumor heterogeneity in breast cancer evolved, and further summarizes recent advances in the detection and monitoring of this heterogeneity in patients with breast cancer.
Collapse
Affiliation(s)
- Jun-Long Song
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Jing-Ping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Sheng-Rong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| |
Collapse
|
17
|
Molecular Ultrasound Imaging of Tissue Inflammation Using an Animal Model of Acute Kidney Injury. Mol Imaging Biol 2016; 17:786-92. [PMID: 25905474 DOI: 10.1007/s11307-015-0860-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE The objective of this study was to evaluate the use of molecular ultrasound (US) imaging for monitoring the early inflammatory effects following acute kidney injury. PROCEDURES A population of rats underwent 30 min of renal ischemia (acute kidney injury, N = 6) or sham injury (N = 4) using established surgical methods. Animals were divided and molecular US imaging was performed during the bolus injection of a targeted microbubble (MB) contrast agent to either P-selectin or vascular cell adhesion molecule 1 (VCAM-1). Imaging was performed before surgery and 4 and 24 h thereafter. After manual segmentation of renal tissue space, the molecular US signal was calculated as the difference between time-intensity curve data before MB injection and after reaching steady-state US image enhancement. All animals were terminated after the 24 h imaging time point and kidneys excised for immunohistochemical (IHC) analysis. RESULTS Renal inflammation was analyzed using molecular US imaging. While results using the P-selectin and VCAM-1 targeted MBs were comparable, it appears that the former was more sensitive to biomarker expression. All molecular US imaging measures had a positive correlation with IHC findings. CONCLUSIONS Acute kidney injury is a serious disease in need of improved noninvasive methods to help diagnose the extent of injury and monitor the tissue throughout disease progression. Molecular US imaging appears well suited to address this challenge and more research is warranted.
Collapse
|
18
|
Three-dimensional Contrast-enhanced Ultrasound in Response Assessment for Breast Cancer: A Comparison with Dynamic Contrast-enhanced Magnetic Resonance Imaging and Pathology. Sci Rep 2016; 6:33832. [PMID: 27652518 PMCID: PMC5031978 DOI: 10.1038/srep33832] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/02/2016] [Indexed: 01/11/2023] Open
Abstract
To compare the capabilities of three-dimensional contrast enhanced ultrasound (3D-CEUS) and dynamic contrast-enhanced magnetic resonance (DCE-MRI) in predicting the response to neoadjuvant chemotherapy (NAC) among breast cancer patients, 48 patients with unilateral breast cancer were recruited for 3D-CEUS and DCE-MRI examinations both before and after NAC; pathology was used to validate the results. This study was approved by the institutional review board, and written informed consent was obtained from each patient. Imaging feature changes and pathological vascularity response, including microvessel density (MVD) and vascular endothelial growth factor (VEGF), were calculated. Pathological complete response (pCR) and major histological response (MHR) were used as references. The 3D-CEUS score, DCE-MRI score, MVD and VEGF significantly decreased (P < 0.0001) after NAC. The correlations between Δ3D-CEUS and ΔDCE-MRI with pCR (r = 0.649, P < 0.0001; r = 0.639, P < 0.0001) and MHR (r = 0.863, P < 0.0001; r = 0.836, P < 0.0001) were significant. All scores showed significant differences between the pCR and non-pCR groups with folder changes of 0.1, 0.1, 2.4, and 2.3, respectively (P = 0.0001, <0.0001, <0.0001 and <0.0001). In conclusion, 3D-CEUS is effective in assessing the response of breast cancer patients undergoing NAC.
Collapse
|
19
|
Abstract
BACKGROUND Contrast-enhanced ultrasound imaging is increasingly being used in clinical applications, particularly for cardiovascular and liver diagnostics. In this context the availability of new molecular contrast agents and the initiation of clinical translation promises new options for pathomechanistic diagnostics. MATERIAL AND METHODS Analysis of the current literature on the development of molecular ultrasound contrast agents, the detection methods as well as the applications in preclinical and clinical studies. RESULTS Molecular contrast agents have become established in preclinical research for the detection of inflammation and angiogenesis and have been continuously refined over recent years. They consist of gas filled microbubbles with a diameter of 1-5 µm and the gas core is stabilized by a shell made of lipids, proteins or polymers to which biomolecules are conjugated that determine the target specificity. The agent BR55 is the first clinically evaluated molecular ultrasound contrast agent. It binds to the angiogenesis marker vascular endothelial growth factor receptor 2 (VEGFR2) and has been studied in several preclinical and clinical phase I and II studies on tumor diagnostics and characterization. CONCLUSION Molecular ultrasound imaging is rapidly evolving in preclinical research for a broad field of applications. Translation to clinical practice is conceivable for many indications and is already ongoing for BR55.
Collapse
Affiliation(s)
- A Rix
- Institut für Experimentelle Molekulare Bildgebung, Pauwelsstrasse 30, 52074, Aachen, Deutschland
| | - M Palmowski
- Institut für Experimentelle Molekulare Bildgebung, Pauwelsstrasse 30, 52074, Aachen, Deutschland
| | - F Kiessling
- Institut für Experimentelle Molekulare Bildgebung, Pauwelsstrasse 30, 52074, Aachen, Deutschland.
| |
Collapse
|
20
|
Wang H, Lutz AM, Hristov D, Tian L, Willmann JK. Intra-Animal Comparison between Three-dimensional Molecularly Targeted US and Three-dimensional Dynamic Contrast-enhanced US for Early Antiangiogenic Treatment Assessment in Colon Cancer. Radiology 2016; 282:443-452. [PMID: 27490690 DOI: 10.1148/radiol.2016160032] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Purpose To perform an intra-animal comparison between (a) three-dimensional (3D) molecularly targeted ultrasonography (US) by using clinical-grade vascular endothelial growth factor receptor 2 (VEGFR2)-targeted microbubbles and (b) 3D dynamic contrast material-enhanced (DCE) US by using nontargeted microbubbles for assessment of antiangiogenic treatment effects in a murine model of human colon cancer. Materials and Methods Twenty-three mice with human colon cancer xenografts were randomized to receive either single-dose antiangiogenic treatment (bevacizumab, n = 14) or control treatment (saline, n = 9). At baseline and 24 hours after treatment, animals were imaged with a clinical US system equipped with a clinical matrix array transducer by using the following techniques: (a) molecularly targeted US with VEGFR2-targeted microbubbles, (b) bolus DCE US with nontargeted microbubbles, and (c) destruction-replenishment DCE US with nontargeted microbubbles. VEGFR2-targeted US signal, peak enhancement, area under the time-intensity curve, time to peak, relative blood volume (rBV), relative blood flow, and blood flow velocity were quantified. VEGFR2 expression and percentage area of blood vessels were assessed ex vivo with quantitative immunofluorescence and correlated with corresponding in vivo US parameters. Statistical analysis was performed with Wilcoxon signed rank tests and rank sum tests, as well as Pearson correlation analysis. Results Molecularly targeted US signal with VEGFR2-targeted microbubbles, peak enhancement, and rBV significantly decreased (P ≤ .03) after a single antiangiogenic treatment compared with those in the control group; similarly, ex vivo VEGFR2 expression (P = .03) and percentage area of blood vessels (P = .03) significantly decreased after antiangiogenic treatment. Three-dimensional molecularly targeted US signal correlated well with VEGFR2 expression (r = 0.86, P = .001), and rBV (r = 0.71, P = .01) and relative blood flow (r = 0.78, P = .005) correlated well with percentage area of blood vessels, while other US perfusion parameters did not. Conclusion Three-dimensional molecularly targeted US and destruction-replenishment 3D DCE US provide complementary molecular and functional in vivo imaging information on antiangiogenic treatment effects in human colon cancer xenografts compared with ex vivo reference standards. © RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Huaijun Wang
- From the Department of Radiology and Molecular Imaging Program at Stanford (H.W., A.M.L., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), School of Medicine, Stanford University, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Amelie M Lutz
- From the Department of Radiology and Molecular Imaging Program at Stanford (H.W., A.M.L., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), School of Medicine, Stanford University, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Dimitre Hristov
- From the Department of Radiology and Molecular Imaging Program at Stanford (H.W., A.M.L., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), School of Medicine, Stanford University, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Lu Tian
- From the Department of Radiology and Molecular Imaging Program at Stanford (H.W., A.M.L., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), School of Medicine, Stanford University, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Jürgen K Willmann
- From the Department of Radiology and Molecular Imaging Program at Stanford (H.W., A.M.L., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), School of Medicine, Stanford University, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| |
Collapse
|
21
|
Hu Q, Wang XY, Kang LK, Wei HM, Xu CM, Wang T, Wen ZH. RGD-Targeted Ultrasound Contrast Agent for Longitudinal Assessment of Hep-2 Tumor Angiogenesis In Vivo. PLoS One 2016; 11:e0149075. [PMID: 26862757 PMCID: PMC4749330 DOI: 10.1371/journal.pone.0149075] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/26/2016] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE To prepare arginine-glycine-aspartate (RGD)-targeted ultrasound contrast microbubbles (MBs) and explore the feasibility of their use in assessing dynamic changes in αvβ3 integrin expression in a murine model of tumor angiogenesis. METHODS RGD peptides were conjugated to the surfaces of microbubbles via biotin-avidin linkage. Microbubbles bearing RADfK peptides were prepared as controls. The RGD-MBs were characterized using an Accusizer 780 and optical microscopy. The binding specificity of the RGD-MBs for ανβ3-expressing endothelial cells (bEnd.3) was demonstrated in vitro by a competitive inhibition experiment. In an in vivo study, mice bearing tumors of three different stages were intravenously injected with RGD-MBs and subjected to targeted, contrast-enhanced, high-frequency ultrasound. Subsequently, tumors were harvested and sectioned for immunofluorescence analysis of ανβ3 expression. RESULTS The mean size of the RGD-MBs was 2.36 ± 1.7 μm. The RGD-MBs showed significantly higher adhesion levels to bEnd.3 cells compared to control MBs (P < 0.01). There was rarely binding of RGD-MBs to αvβ3-negative MCF-7 cells. Adhesion of the RGD-MBs to the bEnd.3 cells was significantly inhibited following treatment with anti-alpha(v) antibodies. The quantitative acoustic video intensity for high-frequency, contrast-enhanced ultrasound imaging of subcutaneous human laryngeal carcinoma (Hep-2) tumor xenografts was significantly higher in small tumors (19.89 ± 2.49) than in medium tumors (11.25 ± 2.23) and large tumors (3.38 ± 0.67) (P < 0.01). CONCLUSIONS RGD-MBs enable noninvasive in vivo visualization of changes in tumor angiogenesis during tumor growth in subcutaneous cancer xenografts.
Collapse
Affiliation(s)
- Qiao Hu
- Department of Diagnostic Ultrasound, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- * E-mail:
| | - Xiao-Yan Wang
- Department of Diagnostic Ultrasound, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Li-Ke Kang
- Department of Diagnostic Ultrasound, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Hai-Ming Wei
- Department of Pathology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Chun-Mei Xu
- Department of Diagnostic Ultrasound, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Tao Wang
- Department of Otolaryngology-Head & Neck Surgery, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zong-Hua Wen
- Department of Pathology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
22
|
Three-dimensional ultrasound molecular imaging of angiogenesis in colon cancer using a clinical matrix array ultrasound transducer. Invest Radiol 2015; 50:322-9. [PMID: 25575176 DOI: 10.1097/rli.0000000000000128] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES We sought to assess the feasibility and reproducibility of 3-dimensional ultrasound molecular imaging (USMI) of vascular endothelial growth factor receptor 2 (VEGFR2) expression in tumor angiogenesis using a clinical matrix array transducer and a clinical grade VEGFR2-targeted contrast agent in a murine model of human colon cancer. MATERIALS AND METHODS Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care. Mice with human colon cancer xenografts (n = 33) were imaged with a clinical ultrasound system and transducer (Philips iU22; X6-1) after intravenous injection of either clinical grade VEGFR2-targeted microbubbles or nontargeted control microbubbles. Nineteen mice were scanned twice to assess imaging reproducibility. Fourteen mice were scanned both before and 24 hours after treatment with either bevacizumab (n = 7) or saline only (n = 7). Three-dimensional USMI data sets were retrospectively reconstructed into multiple consecutive 1-mm-thick USMI data sets to simulate 2-dimensional imaging. Vascular VEGFR2 expression was assessed ex vivo using immunofluorescence. RESULTS Three-dimensional USMI was highly reproducible using both VEGFR2-targeted microbubbles and nontargeted control microbubbles (intraclass correlation coefficient, 0.83). The VEGFR2-targeted USMI signal significantly (P = 0.02) decreased by 57% after antiangiogenic treatment compared with the control group, which correlated well with ex vivo VEGFR2 expression on immunofluorescence (ρ = 0.93, P = 0.003). If only central 1-mm tumor planes were analyzed to assess antiangiogenic treatment response, the USMI signal change was significantly (P = 0.006) overestimated by an average of 27% (range, 2%-73%) compared with 3-dimensional USMI. CONCLUSIONS Three-dimensional USMI is feasible and highly reproducible and allows accurate assessment and monitoring of VEGFR2 expression in tumor angiogenesis in a murine model of human colon cancer.
Collapse
|
23
|
Nakata N, Ohta T, Nishioka M, Takeyama H, Toriumi Y, Kato K, Nogi H, Kamio M, Fukuda K. Optimization of Region of Interest Drawing for Quantitative Analysis: Differentiation Between Benign and Malignant Breast Lesions on Contrast-Enhanced Sonography. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2015; 34:1969-1976. [PMID: 26384607 DOI: 10.7863/ultra.14.10042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/07/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVES This study was performed to evaluate the diagnostic utility of quantitative analysis of benign and malignant breast lesions using contrast-enhanced sonography. METHODS Contrast-enhanced sonography using the perflubutane-based contrast agent Sonazoid (Daiichi Sankyo, Tokyo, Japan) was performed in 94 pathologically proven palpable breast mass lesions, which could be depicted with B-mode sonography. Quantitative analyses using the time-intensity curve on contrast-enhanced sonography were performed in 5 region of interest (ROI) types (manually traced ROI and circular ROIs of 5, 10, 15, and 20 mm in diameter). The peak signal intensity, initial slope, time to peak, positive enhancement integral, and wash-out ratio were investigated in each ROI. RESULTS There were significant differences between benign and malignant lesions in the time to peak (P < .05), initial slope (P < .001), and positive enhancement integral (P < .05) for the manual ROI. Significant differences were found between benign and malignant lesions in the time to peak (P < .05) for the 5-mm ROI; the time to peak (P < .05) and initial slope (P< .05) for the 10-mm ROI; absolute values of the peak signal intensity (P< .05), time to peak (P< .01), and initial slope (P< .005) for the 15-mm ROI; and the time to peak (P < .05) and initial slope (P < .05) for the 20-mm ROI. There were no statistically significant differences in any wash-out ratio values for the 5 ROI types. CONCLUSIONS Kinetic analysis using contrast-enhanced sonography is useful for differentiation between benign and malignant breast lesions.
Collapse
Affiliation(s)
- Norio Nakata
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan.
| | - Tomoyuki Ohta
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan
| | - Makiko Nishioka
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan
| | - Hiroshi Takeyama
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan
| | - Yasuo Toriumi
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan
| | - Kumiko Kato
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan
| | - Hiroko Nogi
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan
| | - Makiko Kamio
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan
| | - Kunihiko Fukuda
- Department of Radiology, Jikei University, School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Hoyt K, Umphrey H, Lockhart M, Robbin M, Forero-Torres A. Ultrasound imaging of breast tumor perfusion and neovascular morphology. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:2292-302. [PMID: 26116159 PMCID: PMC4526459 DOI: 10.1016/j.ultrasmedbio.2015.04.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 03/17/2015] [Accepted: 04/23/2015] [Indexed: 05/09/2023]
Abstract
A novel image processing strategy is detailed for simultaneous measurement of tumor perfusion and neovascular morphology parameters from a sequence of dynamic contrast-enhanced ultrasound (DCE-US) images. After normalization and tumor segmentation, a global time-intensity curve describing contrast agent flow was analyzed to derive surrogate measures of tumor perfusion (i.e., peak intensity, time-to-peak intensity, area under the curve, wash-in rate, wash-out rate). A maximum intensity image was generated from these same segmented image sequences, and each vascular component was skeletonized via a thinning algorithm. This skeletonized data set and collection of vessel segments were then investigated to extract parameters related to the neovascular network and physical architecture (i.e., vessel-to-tissue ratio, number of bifurcations, vessel count, average vessel length and tortuosity). An efficient computation of local perfusion parameters was also introduced and operated by averaging time-intensity curve data over each individual neovascular segment. Each skeletonized neovascular segment was then color-coded by these local measures to produce a parametric map detailing spatial properties of tumor perfusion. Longitudinal DCE-US image data sets were collected in six patients diagnosed with invasive breast cancer using a Philips iU22 ultrasound system equipped with a L9-3 transducer and Definity contrast agent. Patients were imaged using US before and after contrast agent dosing at baseline and again at weeks 6, 12, 18 and 24 after treatment started. Preliminary clinical results suggested that breast tumor response to neoadjuvant chemotherapy may be associated with temporal and spatial changes in DCE-US-derived parametric measures of tumor perfusion. Moreover, changes in neovascular morphology parametric measures may also help identify any breast tumor response (or lack thereof) to systemic treatment. Breast cancer management from early detection to therapeutic monitoring is currently undergoing profound changes. Novel imaging techniques that are sensitive to the unique biological conditions of each individual tumor represent valuable tools in the pursuit of personalized medicine.
Collapse
Affiliation(s)
- Kenneth Hoyt
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | - Heidi Umphrey
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mark Lockhart
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michelle Robbin
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andres Forero-Torres
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
25
|
Yeh JSM, Sennoga CA, McConnell E, Eckersley R, Tang MX, Nourshargh S, Seddon JM, Haskard DO, Nihoyannopoulos P. Quantitative ultrasound molecular imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:2478-2496. [PMID: 26044707 DOI: 10.1016/j.ultrasmedbio.2015.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/10/2015] [Accepted: 04/21/2015] [Indexed: 06/04/2023]
Abstract
Ultrasound molecular imaging using targeting microbubbles is predominantly a semi-quantitative tool, thus limiting its potential diagnostic power and clinical applications. In the work described here, we developed a novel method for acoustic quantification of molecular expression. E-Selectin expression in the mouse heart was induced by lipopolysaccharide. Real-time ultrasound imaging of E-selectin expression in the heart was performed using E-selectin-targeting microbubbles and a clinical ultrasound scanner in contrast pulse sequencing mode at 14 MHz, with a mechanical index of 0.22-0.26. The level of E-selectin expression was quantified using a novel time-signal intensity curve analytical method based on bubble elimination, which consisted of curve-fitting the bi-exponential equation [Formula: see text] to the elimination phase of the myocardial time-signal intensity curve. Ar and Af represent the maximum signal intensities of the retained and freely circulating bubbles in the myocardium, respectively; λr and λf represent the elimination rate constants of the retained and freely circulating bubbles in the myocardium, respectively. Ar correlated strongly with the level of E-selectin expression (|r|>0.8), determined using reverse transcriptase real-time quantitative polymerase chain reaction, and the duration of post-lipopolysaccharide treatment-both linearly related to cell surface E-selectin protein (actual bubble target) concentration in the expression range imaged. Compared with a conventional acoustic quantification method (which used retained bubble signal intensity at 20 min post-bubble injection), this new approach exhibited greater dynamic range and sensitivity and was able to simultaneously quantify other useful characteristics (e.g., the microbubble half-life). In conclusion, quantitative determination of the level of molecular expression is feasible acoustically using a time-signal intensity curve analytical method based on bubble elimination.
Collapse
Affiliation(s)
- James Shue-Min Yeh
- National Heart and Lung Institute, Imperial College London, London, UK; Department of Cardiology, Hammersmith Hospital, London, UK; Imaging Sciences Department, Medical Research Council, Imperial College London, London, UK
| | - Charles A Sennoga
- Imaging Sciences Department, Medical Research Council, Imperial College London, London, UK; Department of Chemistry, Imperial College London, London, UK
| | - Ellen McConnell
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Robert Eckersley
- Imaging Sciences Department, Medical Research Council, Imperial College London, London, UK
| | - Meng-Xing Tang
- Department of Bioengineering, Imperial College London, London, UK
| | - Sussan Nourshargh
- National Heart and Lung Institute, Imperial College London, London, UK; William Harvey Research Institute, Queen Mary, University of London, London, UK
| | - John M Seddon
- Department of Chemistry, Imperial College London, London, UK
| | - Dorian O Haskard
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Petros Nihoyannopoulos
- National Heart and Lung Institute, Imperial College London, London, UK; Department of Cardiology, Hammersmith Hospital, London, UK.
| |
Collapse
|
26
|
Kurihara H, Shimizu C, Miyakita Y, Yoshida M, Hamada A, Kanayama Y, Yonemori K, Hashimoto J, Tani H, Kodaira M, Yunokawa M, Yamamoto H, Watanabe Y, Fujiwara Y, Tamura K. Molecular imaging using PET for breast cancer. Breast Cancer 2015; 23:24-32. [PMID: 25917108 DOI: 10.1007/s12282-015-0613-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/16/2015] [Indexed: 01/27/2023]
Abstract
Molecular imaging can visualize the biological processes at the molecular and cellular levels in vivo using certain tracers for specific molecular targets. Molecular imaging of breast cancer can be performed with various imaging modalities, however, positron emission tomography (PET) is a sensitive and non-invasive molecular imaging technology and this review will focus on PET molecular imaging of breast cancer, such as FDG-PET, FLT-PET, hormone receptor PET, and anti-HER2 PET.
Collapse
Affiliation(s)
- Hiroaki Kurihara
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Chikako Shimizu
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuji Miyakita
- Department of Neurosurgery, National Cancer Center Hospital, Tokyo, Japan
| | - Masayuki Yoshida
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Akinobu Hamada
- Department of Clinical Pharmacology Group for Translational Research Support Core, National Cancer Center Research Institute, Tokyo, Japan
| | | | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Jun Hashimoto
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hitomi Tani
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Makoto Kodaira
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Mayu Yunokawa
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Harukaze Yamamoto
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Yasuhiro Fujiwara
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
27
|
Ultrasound molecular imaging of tumor angiogenesis with a neuropilin-1-targeted microbubble. Biomaterials 2015; 56:104-13. [PMID: 25934284 DOI: 10.1016/j.biomaterials.2015.03.043] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 03/15/2015] [Accepted: 03/20/2015] [Indexed: 02/06/2023]
Abstract
Ultrasound molecular imaging has great potential to impact early disease diagnosis, evaluation of disease progression and the development of target-specific therapy. In this paper, two neuropilin-1 (NRP) targeted peptides, CRPPR and ATWLPPR, were conjugated onto the surface of lipid microbubbles (MBs) to evaluate molecular imaging of tumor angiogenesis in a breast cancer model. Development of a molecular imaging agent using CRPPR has particular importance due to the previously demonstrated internalizing capability of this and similar ligands. In vitro, CRPPR MBs bound to an NRP-expressing cell line 2.6 and 15.6 times more than ATWLPPR MBs and non-targeted (NT) MBs, respectively, and the binding was inhibited by pretreating the cells with an NRP antibody. In vivo, the backscattered intensity within the tumor, relative to nearby vasculature, increased over time during the ∼6 min circulation of the CRPPR-targeted contrast agents providing high contrast images of angiogenic tumors. Approximately 67% of the initial signal from CRPPR MBs remained bound after the majority of circulating MBs had cleared (8 min), 8 and 4.5 times greater than ATWLPPR and NT MBs, respectively. Finally, at 7-21 days after the first injection, we found that CRPPR MBs cleared faster from circulation and tumor accumulation was reduced likely due to a complement-mediated recognition of the targeted microbubble and a decrease in angiogenic vasculature, respectively. In summary, we find that CRPPR MBs specifically bind to NRP-expressing cells and provide an effective new agent for molecular imaging of angiogenesis.
Collapse
|
28
|
El Kaffas A, Czarnota GJ. Biomechanical effects of microbubbles: from radiosensitization to cell death. Future Oncol 2015; 11:1093-108. [DOI: 10.2217/fon.15.19] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
ABSTRACT Ultrasound-stimulated microbubbles have been demonstrated to mechanically perturb cell membranes, resulting in the activation of biological signaling pathways that significantly enhance the effects of radiation. The underlying mechanism involves augmented ceramide production following both microbubble stimulation and irradiation, leading to rapid and extensive endothelial apoptosis and tumor cell death as a result of vascular collapse. Endothelial cells are particularly sensitive to ceramide-induced cell death due to an enriched presence of sphingomyelinase in their membranes. In tumors, this consequent rapid vascular shutdown translates to an overall increase in tumor responses to radiation treatments. This review summarizes the groundwork behind endothelial-based radiation enhancement with ultrasound-stimulated microbubbles, and presents ongoing research on the use of microbubbles as therapeutic agents in cancer therapy.
Collapse
Affiliation(s)
- Ahmed El Kaffas
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Imaging Research & Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Gregory J Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Imaging Research & Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
29
|
Wu Y, Peng H, Zhao X. Diagnostic performance of contrast-enhanced ultrasound for ovarian cancer: a meta-analysis. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:967-974. [PMID: 25701533 DOI: 10.1016/j.ultrasmedbio.2014.11.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/08/2014] [Accepted: 11/24/2014] [Indexed: 06/04/2023]
Abstract
This meta-analysis is the first study aimed at assessing the overall diagnostic performance of contrast-enhanced ultrasound for ovarian cancer. PubMed, Embase and Medline databases were systematically searched for relevant articles published up to June 2014. Data were pooled to yield summary sensitivity, specificity, diagnostic odds ratio and receiver operating characteristic curves using Meta-Disc Version 1.4 software. Ten independent studies with 579 ovarian tumors were enrolled in this meta-analysis. The pooled sensitivity, specificity and diagnostic odds ratio statistics were 0.89 (0.83-0.94), 0.91 (0.88-0.93) and 91.70 (41.41-203.05), respectively, and the area under the summary receiver operating characteristic curve was 0.9619 (standard error: 0.0125), all indicating that contrast-enhanced ultrasound has high diagnostic accuracy in differentiation of malignant from benign ovarian tumors.
Collapse
Affiliation(s)
- Ying Wu
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Hongling Peng
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
30
|
Warram JM, de Boer E, Sorace AG, Chung TK, Kim H, Pleijhuis RG, van Dam GM, Rosenthal EL. Antibody-based imaging strategies for cancer. Cancer Metastasis Rev 2015; 33:809-22. [PMID: 24913898 DOI: 10.1007/s10555-014-9505-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although mainly developed for preclinical research and therapeutic use, antibodies have high antigen specificity, which can be used as a courier to selectively deliver a diagnostic probe or therapeutic agent to cancer. It is generally accepted that the optimal antigen for imaging will depend on both the expression in the tumor relative to normal tissue and the homogeneity of expression throughout the tumor mass and between patients. For the purpose of diagnostic imaging, novel antibodies can be developed to target antigens for disease detection, or current FDA-approved antibodies can be repurposed with the covalent addition of an imaging probe. Reuse of therapeutic antibodies for diagnostic purposes reduces translational costs since the safety profile of the antibody is well defined and the agent is already available under conditions suitable for human use. In this review, we will explore a wide range of antibodies and imaging modalities that are being translated to the clinic for cancer identification and surgical treatment.
Collapse
Affiliation(s)
- Jason M Warram
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abou-Elkacem L, Bachawal SV, Willmann JK. Ultrasound molecular imaging: Moving toward clinical translation. Eur J Radiol 2015; 84:1685-93. [PMID: 25851932 DOI: 10.1016/j.ejrad.2015.03.016] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/13/2015] [Indexed: 12/11/2022]
Abstract
Ultrasound is a widely available, cost-effective, real-time, non-invasive and safe imaging modality widely used in the clinic for anatomical and functional imaging. With the introduction of novel molecularly-targeted ultrasound contrast agents, another dimension of ultrasound has become a reality: diagnosing and monitoring pathological processes at the molecular level. Most commonly used ultrasound molecular imaging contrast agents are micron sized, gas-containing microbubbles functionalized to recognize and attach to molecules expressed on inflamed or angiogenic vascular endothelial cells. There are several potential clinical applications currently being explored including earlier detection, molecular profiling, and monitoring of cancer, as well as visualization of ischemic memory in transient myocardial ischemia, monitoring of disease activity in inflammatory bowel disease, and assessment of arteriosclerosis. Recently, a first clinical grade ultrasound contrast agent (BR55), targeted at a molecule expressed in neoangiogenesis (vascular endothelial growth factor receptor type 2; VEGFR2) has been introduced and safety and feasibility of VEGFR2-targeted ultrasound imaging is being explored in first inhuman clinical trials in various cancer types. This review describes the design of ultrasound molecular imaging contrast agents, imaging techniques, and potential future clinical applications of ultrasound molecular imaging.
Collapse
Affiliation(s)
- Lotfi Abou-Elkacem
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, CA, USA
| | - Sunitha V Bachawal
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jürgen K Willmann
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, CA, USA.
| |
Collapse
|
32
|
van Rooij T, Daeichin V, Skachkov I, de Jong N, Kooiman K. Targeted ultrasound contrast agents for ultrasound molecular imaging and therapy. Int J Hyperthermia 2015; 31:90-106. [PMID: 25707815 DOI: 10.3109/02656736.2014.997809] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ultrasound contrast agents (UCAs) are used routinely in the clinic to enhance contrast in ultrasonography. More recently, UCAs have been functionalised by conjugating ligands to their surface to target specific biomarkers of a disease or a disease process. These targeted UCAs (tUCAs) are used for a wide range of pre-clinical applications including diagnosis, monitoring of drug treatment, and therapy. In this review, recent achievements with tUCAs in the field of molecular imaging, evaluation of therapy, drug delivery, and therapeutic applications are discussed. We present the different coating materials and aspects that have to be considered when manufacturing tUCAs. Next to tUCA design and the choice of ligands for specific biomarkers, additional techniques are discussed that are applied to improve binding of the tUCAs to their target and to quantify the strength of this bond. As imaging techniques rely on the specific behaviour of tUCAs in an ultrasound field, it is crucial to understand the characteristics of both free and adhered tUCAs. To image and quantify the adhered tUCAs, the state-of-the-art techniques used for ultrasound molecular imaging and quantification are presented. This review concludes with the potential of tUCAs for drug delivery and therapeutic applications.
Collapse
Affiliation(s)
- Tom van Rooij
- Department of Biomedical Engineering, Thoraxcenter , Erasmus MC, Rotterdam , the Netherlands
| | | | | | | | | |
Collapse
|
33
|
Zhou Y, Gu H, Xu Y, Li F, Kuang S, Wang Z, Zhou X, Ma H, Li P, Zheng Y, Ran H, Jian J, Zhao Y, Song W, Wang Q, Wang D. Targeted antiangiogenesis gene therapy using targeted cationic microbubbles conjugated with CD105 antibody compared with untargeted cationic and neutral microbubbles. Theranostics 2015; 5:399-417. [PMID: 25699099 PMCID: PMC4329503 DOI: 10.7150/thno.10351] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 01/02/2015] [Indexed: 12/03/2022] Open
Abstract
Objective This study aimed to develop targeted cationic microbubbles conjugated with a CD105 antibody (CMB105) for use in targeted vascular endothelial cell gene therapy and ultrasound imaging. We compared the results with untargeted cationic microbubbles (CMB) and neutral microbubbles (NMB). Methods CMB105 were prepared and compared with untargeted CMB and NMB. First, the microbubbles were characterized in terms of size, zeta-potential, antibody binding ability and plasmid DNA loading capacity. A tumor model of subcutaneous breast cancer in nude mice was used for our experiments. The ability of different types of microbubbles to target HUVECs in vitro and tumor neovascularization in vivo was measured. The endostatin gene was selected for its outstanding antiangiogenesis effect. For in vitro experiments, the transfection efficiency and cell cycle were analyzed using flow cytometry, and the transcription and expression of endostatin were measured by qPCR and Western blotting, respectively. Vascular tube cavity formation and tumor cell invasion were used to evaluate the antiangiogenesis gene therapy efficiency in vitro. Tumors were exposed to ultrasound irradiation with different types of microbubbles, and the gene therapy effects were investigated by detecting apoptosis induction and changes in tumor volume. Results CMB105 and CMB differed significantly from NMB in terms of zeta-potential, and the DNA loading capacities were 16.76±1.75 μg, 18.21±1.22 μg, and 0.48±0.04 μg per 5×108 microbubbles, respectively. The charge coupling of plasmid DNA to CMB105 was not affected by the presence of the CD105 antibody. Both CMB105 and CMB could target to HUVECs in vitro, whereas only CMB105 could target to tumor neovascularization in vivo. In in vitro experiments, the transfection efficiency of CMB105 was 24.7-fold higher than the transfection efficiency of NMB and 1.47-fold higher than the transfection efficiency of CMB (P<0.05). With ultrasound-targeted microbubble destruction (UTMD)-mediated gene therapy, the transcription and expression of endostatin were the highest in the CMB105 group (P<0.001); the antiangiogenesis effect and inhibition of tumor cells invasion was better with CMB105 than CMB or NMB in vitro (P<0.01). After gene therapy, the tumor volumes of CMB105 group were significantly smaller than that of CMB and NMB, and many tumor cells had begun apoptosis in the CMB105 group, which had the highest apoptosis index (P<0.001). Conclusions As a contrast agent and plasmid carrier, CMB105 can be used not only for targeted ultrasound imaging but also for targeted gene therapy both in vitro and in vivo. The plasmid DNA binding ability of the CMB was not affected by conjugation of the CMB with the CD105 antibody, and because of its targeting ability, the gene transfection efficiency and therapeutic effect were better compared with the untargeted CMB and NMB. The advantages of targeted gene therapy with CMB105 in vivo were more prominent than with CMB or NMB because neither can target the endothelia in vivo.
Collapse
|
34
|
Mahoney M, Sorace A, Warram J, Samuel S, Hoyt K. Volumetric contrast-enhanced ultrasound imaging of renal perfusion. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2014; 33:1427-37. [PMID: 25063408 PMCID: PMC4135386 DOI: 10.7863/ultra.33.8.1427] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
OBJECTIVES To determine whether volumetric contrast-enhanced ultrasound (US) imaging has the potential to monitor changes in renal perfusion after vascular injury. METHODS Volumetric contrast-enhanced US uses a series of planar image acquisitions, capturing the nonlinear second harmonic signal from microbubble contrast agents flowing in the vasculature. Tissue perfusion parameters (peak intensity [IPK], time to peak intensity [TPK], wash-in rate [WIR], and area under the curve [AUC]) were derived from time-intensity curve data collected during in vitro flow phantom studies and in vivo animal studies of healthy and injured kidneys. For the flow phantom studies, either the contrast agent concentration was held constant (10 μL/L) with varying volumetric flow rates (10, 20, and 30 mL/min), or the flow rate was held constant (30 mL/min) with varying contrast agent concentrations (5, 10, and 20 μL/L). Animal studies used healthy rats or those that underwent renal ischemia-reperfusion injury. Renal studies were performed with healthy rats while the transducer angle was varied for each volumetric contrast-enhanced US image acquisition (reference or 0°, 45°, and 90°) to determine whether repeated renal perfusion measures were isotropic and independent of transducer position. Blood serum biomarkers and immunohistology were used to confirm acute kidney injury. RESULTS Flow phantom results revealed a linear relationship between microbubble concentrations injected into the flow system and the IPK, WIR, and AUC (R(2) > 0.56; P < .005). Furthermore, there was a linear relationship between volume flow rate changes and the TPK, WIR, and AUC (R(2) > 0.77; P < .005). No significant difference was found between the transducer angle during data acquisition and any of the perfusion measures (P > .60). After induction of renal ischemia-reperfusion injury in the rat animal model (n = 4), volumetric contrast-enhanced US imaging of the injured kidney revealed an initial reduction in renal perfusion compared to control animals, followed by progressive recovery of vascular function. CONCLUSIONS Volumetric contrast-enhanced US-based renal perfusion imaging may prove clinically feasible for detecting and monitoring acute kidney injury.
Collapse
Affiliation(s)
- Marshall Mahoney
- Departments of Biomedical Engineering (M.M., A.S.), Radiology (J.W., S.S., K.H.), and Electrical Engineering (K.H.), and Comprehensive Cancer Center (K.H.), University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Anna Sorace
- Departments of Biomedical Engineering (M.M., A.S.), Radiology (J.W., S.S., K.H.), and Electrical Engineering (K.H.), and Comprehensive Cancer Center (K.H.), University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Jason Warram
- Departments of Biomedical Engineering (M.M., A.S.), Radiology (J.W., S.S., K.H.), and Electrical Engineering (K.H.), and Comprehensive Cancer Center (K.H.), University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Sharon Samuel
- Departments of Biomedical Engineering (M.M., A.S.), Radiology (J.W., S.S., K.H.), and Electrical Engineering (K.H.), and Comprehensive Cancer Center (K.H.), University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Kenneth Hoyt
- Departments of Biomedical Engineering (M.M., A.S.), Radiology (J.W., S.S., K.H.), and Electrical Engineering (K.H.), and Comprehensive Cancer Center (K.H.), University of Alabama at Birmingham, Birmingham, Alabama USA.
| |
Collapse
|
35
|
Kiessling F, Fokong S, Bzyl J, Lederle W, Palmowski M, Lammers T. Recent advances in molecular, multimodal and theranostic ultrasound imaging. Adv Drug Deliv Rev 2014; 72:15-27. [PMID: 24316070 DOI: 10.1016/j.addr.2013.11.013] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 11/14/2013] [Accepted: 11/25/2013] [Indexed: 12/12/2022]
Abstract
Ultrasound (US) imaging is an exquisite tool for the non-invasive and real-time diagnosis of many different diseases. In this context, US contrast agents can improve lesion delineation, characterization and therapy response evaluation. US contrast agents are usually micrometer-sized gas bubbles, stabilized with soft or hard shells. By conjugating antibodies to the microbubble (MB) surface, and by incorporating diagnostic agents, drugs or nucleic acids into or onto the MB shell, molecular, multimodal and theranostic MBs can be generated. We here summarize recent advances in molecular, multimodal and theranostic US imaging, and introduce concepts how such advanced MB can be generated, applied and imaged. Examples are given for their use to image and treat oncological, cardiovascular and neurological diseases. Furthermore, we discuss for which therapeutic entities incorporation into (or conjugation to) MB is meaningful, and how US-mediated MB destruction can increase their extravasation, penetration, internalization and efficacy.
Collapse
|
36
|
Warram JM, Sorace AG, Mahoney M, Samuel S, Harbin B, Joshi M, Martin A, Whitworth L, Hoyt K, Zinn KR. Biodistribution of P-selectin targeted microbubbles. J Drug Target 2014; 22:387-94. [PMID: 24731055 DOI: 10.3109/1061186x.2013.869822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To evaluate binding of P-selectin targeted microbubbles (MB) in tumor vasculature; a whole-body imaging and biodistribution study was performed in a tumor bearing mouse model. METHODS Antibodies were radiolabeled with Tc-99 m using the HYNIC method. Tc-99 m labeled anti-P-selectin antibodies were avidin-bound to lipid-shelled, perfluorocarbon gas-filled MB and intravenously injected into mice bearing MDA-MB-231 breast tumors. Whole-body biodistribution was performed at 5 min (n = 12) and 60 min (n = 4) using a gamma counter. Tc-99 m-labeled IgG bound IgG-control-MB group (n = 12 at 5 min; n = 4 at 60 min), Tc-99 m-labeled IgG-control-Ab group (n = 5 at 5 min; n = 3 at 60 min) and Tc-99 m-labeled anti P-selectin-Ab group (n = 5 at 5 min; n = 3 at 60 min) were also evaluated. Planar gamma camera imaging was also performed at each time point. RESULTS Targeted-MB retention in tumor (60 min: 1.8 ± 0.3% ID/g) was significantly greater (p = 0.01) than targeted-MB levels in adjacent skeletal muscle at both time points (5 min: 0.7 ± 0.2% ID/g; 60 min: 0.2 ± 0.1% ID/g) while there was no significant difference (p = 0.17) between muscle and tumor retention for the IgG-control-MB group at 5 min. CONCLUSIONS P-selectin targeted MBs were significantly higher in tumor tissue, as compared with adjacent skeletal tissue or tumor retention of IgG-control-MB.
Collapse
|
37
|
Wei X, Li Y, Zhang S, Gao X, Luo Y, Gao M. Ultrasound targeted apoptosis imaging in monitoring early tumor response of trastuzumab in a murine tumor xenograft model of her-2-positive breast cancer(1.). Transl Oncol 2014; 7:284-91. [PMID: 24685547 PMCID: PMC4101340 DOI: 10.1016/j.tranon.2014.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/05/2014] [Accepted: 02/06/2014] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Our study aimed to monitor the trastuzumab therapy response of murine tumor xenograft model with human epidermal growth factor receptor 2 (Her-2)-positive breast cancer using ultrasound targeted apoptosis imaging. METHODS We prepared targeted apoptosis ultrasound probes by nanobubble (NB) binding with Annexin V. In vitro, we investigated the binding rate of NB-Annexin V with breast cancer apoptotic cells after the trastuzumab treatment. In vivo, tumor-bearing mice underwent ultrasound targeted imaging over 7 days. After imaging was completed, the tumors were excised to determine Her-2 and caspase-3 expression by immunohistochemistry (IHC). The correlation between parameters of imaging and histologic results was then analyzed. RESULTS For seeking the ability of targeted NB binding with apoptotic tumor cells (Her-2 positive), we found that binding rate in the treatment group was higher than that of the control group in vitro (P = .001). There were no differences of tumor sizes in all groups over the treatment process in vivo (P = .98). However, when using ultrasound imaging to visualize tumors by targeted NB in vivo, we observed that the mean and peak intensities from NBs gradually increased in the treatment group after trastuzumab therapy (P = .001). Furthermore, these two parameters were significantly associated with caspase-3 expression of tumor excised samples (P = .0001). CONCLUSION Ultrasound targeted apoptosis imaging can be a non-invasive technique to evaluate the early breast tumor response to trastuzumab therapy.
Collapse
Affiliation(s)
- Xi Wei
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ying Li
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Sheng Zhang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiujun Gao
- Institute of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Yi Luo
- Department of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ming Gao
- Department of Thyroid and Cervical Tumor, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
38
|
Saini R, Hoyt K. Recent developments in dynamic contrast-enhanced ultrasound imaging of tumor angiogenesis. ACTA ACUST UNITED AC 2014; 6:41-52. [PMID: 25221623 DOI: 10.2217/iim.13.74] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Angiogenesis is a critical process for tumor growth and metastatic dissemination. There is tremendous interest in the development of noninvasive methods for imaging tumor angiogenesis, and ultrasound (US) is an emerging platform technology to address this challenge. The introduction of intravascular microbubble contrast agents not only allows real-time visualization of tumor perfusion during an US examination, but they can be functionalized with specific ligands to permit molecular US imaging of angiogenic biomarkers that are overexpressed on the tumor endothelium. In this article, we will review current concepts and developing trends for US imaging of tumor angiogenesis, including relevant preclinical and clinicsal findings.
Collapse
Affiliation(s)
- Reshu Saini
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA ; Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth Hoyt
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA ; Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA ; Electrical & Computer Engineering, University of Alabama at Birmingham, Birmingham, AL, USA ; Comprehensive Cancer Center, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL 35294, USA
| |
Collapse
|
39
|
Mancini M, Greco A, Salvatore G, Liuzzi R, Di Maro G, Vergara E, Chiappetta G, Pasquinelli R, Brunetti A, Salvatore M. Imaging of thyroid tumor angiogenesis with microbubbles targeted to vascular endothelial growth factor receptor type 2 in mice. BMC Med Imaging 2013. [PMID: 24028408 DOI: 0.1186/1471-2342-13-31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND To evaluate whether Contrast Enhanced Ultrasund (CEUS) with microbubbles (MBs) targeted to VEGFR-2 is able to characterize in vivo the VEGFR-2 expression in the tumor vasculature of a mouse model of thyroid cancer (Tg-TRK-T1). METHODS Animal protocol was approved by Institutional committee on Laboratory Animal Care. Contrast-enhanced ultrasound imaging with MBs targeted with an anti-VEGFR-2 monoclonal antibody (UCAVEGFR-2) and isotype control antibody (UCAIgG) was performed in 7 mice with thyroid carcinoma, 5 mice with hyperplasia or benign thyroid nodules and 4 mice with normal thyroid. After ultrasonography, the tumor samples were harvested for histological examination and VEGFR-2 expression was tested by immunohistochemistry. Data were reported as median and range. Paired non parametric Wilcoxon's test and ANOVA of Kruskal-Wallis were used. The correlation between the contrast signal and the VEGFR-2 expression was assessed by the Spearman coefficient. RESULTS The Video intensity difference (VID) caused by backscatter of the retained UCAVEGFR-2 was significantly higher in mice harboring thyroid tumors compared to mice with normal thyroids (P < 0.01) and to mice harboring benign nodules (P < 0.01). No statistically significant differences of VID were observed in the group of mice carrying benign nodules compared to mice with normal thyroids. Moreover in thyroid tumors VID of retained VEGFR-2-targeted UCA was significantly higher than that of control UCAIgG (P <0.05). Results of immunohistochemical analysis confirmed VEGFR-2 overexpression. The magnitude of the molecular ultrasonographic signal from a VEGFR-2-targeted UCA retained by tissue correlates with VEGFR-2 expression determined by immunohistochemistry (rho 0.793, P=0.0003). CONCLUSIONS We demonstrated that CEUS with UCAVEGFR-2 might be used for in vivo non invasive detection and quantification of VEGFR-2 expression in thyroid cancer in mice, and to differentiate benign from malignant thyroid nodules.
Collapse
Affiliation(s)
- Marcello Mancini
- Institute of Biostructure and Bioimaging, Italian National Research Council (CNR), Naples, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mancini M, Greco A, Salvatore G, Liuzzi R, Di Maro G, Vergara E, Chiappetta G, Pasquinelli R, Brunetti A, Salvatore M. Imaging of thyroid tumor angiogenesis with microbubbles targeted to vascular endothelial growth factor receptor type 2 in mice. BMC Med Imaging 2013; 13:31. [PMID: 24028408 PMCID: PMC3848463 DOI: 10.1186/1471-2342-13-31] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 09/06/2013] [Indexed: 12/30/2022] Open
Abstract
Background To evaluate whether Contrast Enhanced Ultrasund (CEUS) with microbubbles (MBs) targeted to VEGFR-2 is able to characterize in vivo the VEGFR-2 expression in the tumor vasculature of a mouse model of thyroid cancer (Tg-TRK-T1). Methods Animal protocol was approved by Institutional committee on Laboratory Animal Care. Contrast-enhanced ultrasound imaging with MBs targeted with an anti-VEGFR-2 monoclonal antibody (UCAVEGFR-2) and isotype control antibody (UCAIgG) was performed in 7 mice with thyroid carcinoma, 5 mice with hyperplasia or benign thyroid nodules and 4 mice with normal thyroid. After ultrasonography, the tumor samples were harvested for histological examination and VEGFR-2 expression was tested by immunohistochemistry. Data were reported as median and range. Paired non parametric Wilcoxon’s test and ANOVA of Kruskal-Wallis were used. The correlation between the contrast signal and the VEGFR-2 expression was assessed by the Spearman coefficient. Results The Video intensity difference (VID) caused by backscatter of the retained UCAVEGFR-2 was significantly higher in mice harboring thyroid tumors compared to mice with normal thyroids (P < 0.01) and to mice harboring benign nodules (P < 0.01). No statistically significant differences of VID were observed in the group of mice carrying benign nodules compared to mice with normal thyroids. Moreover in thyroid tumors VID of retained VEGFR-2-targeted UCA was significantly higher than that of control UCAIgG (P <0.05). Results of immunohistochemical analysis confirmed VEGFR-2 overexpression. The magnitude of the molecular ultrasonographic signal from a VEGFR-2-targeted UCA retained by tissue correlates with VEGFR-2 expression determined by immunohistochemistry (rho 0.793, P=0.0003). Conclusions We demonstrated that CEUS with UCAVEGFR-2 might be used for in vivo non invasive detection and quantification of VEGFR-2 expression in thyroid cancer in mice, and to differentiate benign from malignant thyroid nodules.
Collapse
Affiliation(s)
- Marcello Mancini
- Institute of Biostructure and Bioimaging, Italian National Research Council (CNR), Naples, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Streeter JE, Dayton PA. An in vivo evaluation of the effect of repeated administration and clearance of targeted contrast agents on molecular imaging signal enhancement. Theranostics 2013; 3:93-8. [PMID: 23424189 PMCID: PMC3575589 DOI: 10.7150/thno.5341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 11/21/2012] [Indexed: 12/31/2022] Open
Abstract
Competitive inhibition diminishes ligand adhesion as receptor sites become occupied with competing ligands. It is unknown if this effect occurs in ultrasound molecular imaging studies where endothelial binding sites become occupied with adherent bubbles or bubble fragments. The goal of this pilot study was to assess the effect that repeated administration and clearance of targeted agents has on successive adhesion. Two groups of animals were imaged with 3-D ultrasonic molecular imaging. Injections and imaging were performed on Group 1 at time 0 and 60 minutes. Group 2 received injections of microbubbles at 0, 15, 30, 45 and 60 minutes with imaging at 0 and 60 minutes. At 60 minutes, Group 1 targeting relative to baseline was not significantly different from Group 2 (1.06±0.27 vs. 1.08±0.34, p=0.93). Data suggest that multiple injections of targeted microbubbles do not block sufficient binding sites to bias molecular imaging data in serial studies.
Collapse
|