1
|
Beckman RA. Neutral evolution of rare cancer mutations in the computer and the clinic. NPJ Syst Biol Appl 2024; 10:110. [PMID: 39358357 PMCID: PMC11447017 DOI: 10.1038/s41540-024-00436-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
A distinct model of neutral evolution of rare cancer mutations is described and contrasted with models relying on the infinite sites approximation (that a specific mutation arises in only one cell at any instant). An explosion of genetic diversity is predicted at clinical cell numbers and may explain the progressive refractoriness of cancers during a clinical course. The widely used infinite sites assumption may not be applicable for clinical cancers.
Collapse
Affiliation(s)
- Robert A Beckman
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA.
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
2
|
Trencsényi G, Enyedi KN, Mező G, Halmos G, Képes Z. NGR-Based Radiopharmaceuticals for Angiogenesis Imaging: A Preclinical Review. Int J Mol Sci 2023; 24:12675. [PMID: 37628856 PMCID: PMC10454655 DOI: 10.3390/ijms241612675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Angiogenesis plays a crucial role in tumour progression and metastatic spread; therefore, the development of specific vectors targeting angiogenesis has attracted the attention of several researchers. Since angiogenesis-associated aminopeptidase N (APN/CD13) is highly expressed on the surface of activated endothelial cells of new blood vessels and a wide range of tumour cells, it holds great promise for imaging and therapy in the field of cancer medicine. The selective binding capability of asparagine-glycine-arginine (NGR) motif containing molecules to APN/CD13 makes radiolabelled NGR peptides promising radiopharmaceuticals for the non-invasive, real-time imaging of APN/CD13 overexpressing malignancies at the molecular level. Preclinical small animal model systems are major keystones for the evaluation of the in vivo imaging behaviour of radiolabelled NGR derivatives. Based on existing literature data, several positron emission tomography (PET) and single-photon emission computed tomography (SPECT) radioisotopes have been applied so far for the labelling of tumour vasculature homing NGR sequences such as Gallium-68 (68Ga), Copper-64 (64Cu), Technetium-99m (99mTc), Lutetium-177 (177Lu), Rhenium-188 (188Re), or Bismuth-213 (213Bi). Herein, a comprehensive overview is provided of the recent preclinical experiences with radiolabelled imaging probes targeting angiogenesis.
Collapse
Affiliation(s)
- György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary;
| | - Kata Nóra Enyedi
- ELKH-ELTE Research Group of Peptide Chemistry, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary; (K.N.E.); (G.M.)
- Institute of Chemistry, Faculty of Science, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Gábor Mező
- ELKH-ELTE Research Group of Peptide Chemistry, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary; (K.N.E.); (G.M.)
- Institute of Chemistry, Faculty of Science, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Gábor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary;
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary;
| |
Collapse
|
3
|
Chalfant H, Bonds M, Scott K, Condacse A, Dennahy IS, Martin WT, Little C, Edil BH, McNally LR, Jain A. Innovative Imaging Techniques Used to Evaluate Borderline-Resectable Pancreatic Adenocarcinoma. J Surg Res 2023; 284:42-53. [PMID: 36535118 PMCID: PMC10131671 DOI: 10.1016/j.jss.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 09/15/2022] [Accepted: 10/11/2022] [Indexed: 12/23/2022]
Abstract
A diagnosis of pancreatic cancer carries a 5-y survival rate of less than 10%. Furthermore, the detection of pancreatic cancer occurs most often in later stages of the disease due to its location in the retroperitoneum and lack of symptoms (in most cases) until tumors become more advanced. Once diagnosed, cross-sectional imaging techniques are heavily utilized to determine the tumor stage and the potential for surgical resection. However, a major determinant of resectability is the extent of local vascular involvement of the mesenteric vessels and critical tributaries; current imaging techniques have limited capacity to accurately determine vascular involvement. Surrounding inflammation and fibrosis can be difficult to discriminate from viable tumor, making determination of the degree of vascular involvement unreliable. New innovations in fluorescence and optoacoustic imaging techniques may overcome these limitations and make determination of resectability more accurate. These imaging modalities are able to more clearly discern between viable tumor tissue and non-neoplastic inflammation or desmoplasia, allowing clinicians to more reliably characterize vascular involvement and develop individualized treatment plans for patients. This review will discuss the current imaging techniques used to diagnose pancreatic cancer, the barriers that current techniques raise to accurate staging, and novel fluorescence and optoacoustic imaging techniques that may provide more accurate clinical staging of pancreatic cancer.
Collapse
Affiliation(s)
- Hunter Chalfant
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Morgan Bonds
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Kristina Scott
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Anna Condacse
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Isabel S Dennahy
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - W Taylor Martin
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Cooper Little
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Barish H Edil
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Lacey R McNally
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma.
| | - Ajay Jain
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma.
| |
Collapse
|
4
|
Therapeutic Performance Evaluation of 213Bi-Labelled Aminopeptidase N (APN/CD13)-Affine NGR-Motif ([ 213Bi]Bi-DOTAGA-cKNGRE) in Experimental Tumour Model: A Treasured Tailor for Oncology. Pharmaceutics 2023; 15:pharmaceutics15020491. [PMID: 36839813 PMCID: PMC9968005 DOI: 10.3390/pharmaceutics15020491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Since NGR-tripeptides (asparagine-glycine-arginine) selectively target neoangiogenesis-associated Aminopeptidase N (APN/CD13) on cancer cells, we aimed to evaluate the in vivo tumour targeting capability of radiolabelled, NGR-containing, ANP/CD13-selective [213Bi]Bi-DOTAGA-cKNGRE in CD13pos. HT1080 fibrosarcoma-bearing severe combined immunodeficient CB17 mice. 10 ± 1 days after cancer cell inoculation, positron emission tomography (PET) was performed applying [68Ga]Ga-DOTAGA-cKNGRE for tumour verification. On the 7th, 8th, 10th and 12th days the treated group of tumourous mice were intraperitoneally administered with 4.68 ± 0.10 MBq [213Bi]Bi-DOTAGA-cKNGRE, while the untreated tumour-bearing animals received 150 μL saline solution. In addition to body weight (BW) and tumour volume measurements, ex vivo biodistribution studies were conducted 30 and 90 min postinjection (pi.). The following quantitative standardised uptake values (SUV) confirmed the detectability of the HT1080 tumours: SUVmean and SUVmax: 0.37 ± 0.09 and 0.86 ± 0.14, respectively. Although no significant difference (p ≤ 0.05) was encountered between the BW of the treated and untreated mice, their tumour volumes measured on the 9th, 10th and 12th days differed significantly (p ≤ 0.01). Relatively higher [213Bi]Bi-DOTAGA-cKNGRE accumulation of the HT1080 neoplasms (%ID/g: 0.80 ± 0.16) compared with the other organs at 90 min time point yields better tumour-to-background ratios. Therefore, the therapeutic application of APN/CD13-affine [213Bi]Bi-DOTAGA- cKNGRE seems to be promising in receptor-positive fibrosarcoma treatment.
Collapse
|
5
|
Characterization of spatially mapped volumetric molecular ultrasound signals for predicting response to anti-vascular therapy. Sci Rep 2023; 13:1686. [PMID: 36717575 PMCID: PMC9886917 DOI: 10.1038/s41598-022-26273-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 12/13/2022] [Indexed: 01/31/2023] Open
Abstract
Quantitative three-dimensional molecular ultrasound is a promising technology for longitudinal imaging applications such as therapy monitoring; the risk profile is favorable compared to positron emission tomography and computed tomography. However, clinical translation of quantitative methods for this technology are limited in that they assume that tumor tissues are homogeneous, and often depend on contrast-destruction events that can produce unintended bioeffects. Here, we develop quantitative features (henceforth image features) that capture tumor spatial information, and that are extracted without contrast destruction. We compare these techniques with the contrast-destruction derived differential targeted enhancement parameter (dTE) in predicting response to therapy. We found thirty-three reproducible image features that predict response to antiangiogenic therapy, without the need for a contrast agent disruption pulse. Multiparametric analysis shows that several of these image features can differentiate treated versus control animals with comparable performance to post-destruction measurements, suggesting that these can potentially replace parameters such as the dTE. The highest performing pre-destruction image features showed strong linear correlations with conventional dTE parameters with less overall variance. Thus, our study suggests that image features obtained during the wash in of the molecular agent, pre-destruction, may replace conventional post-destruction image features or the dTE parameter.
Collapse
|
6
|
Mi X, Guo X, Du H, Han M, Liu H, Luo Y, Wang D, Xiang R, Yue S, Zhang Y, Tan X. Combined legumain- and integrin-targeted nanobubbles for molecular ultrasound imaging of breast cancer. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 42:102533. [PMID: 35150904 DOI: 10.1016/j.nano.2022.102533] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/31/2022]
Abstract
Molecular ultrasound imaging is a promising strategy for non-invasive and precise cancer diagnosis. Previously reported ultrasound contrast agents (UCAs) are mostly microbubbles or nanobubbles (NBs) larger than 200 nm, leading to less efficient tumor delivery. Here we synthesized NBs with a small size (~49 nm) and modified the NB surface with alanine-alanine-asparagine (NB-A) or arginine-glycine-aspartic acid peptide (NB-R) for concurrent active targeting towards legumain in tumor cells and integrin in tumor neovasculature. In vitro, the NB-A and NB-R presented echogenicity comparable with SonoVue MBs and showed specific binding with tumors cells and endothelial cells, respectively. In vivo, the combined NB-A/NB-R accumulated in tumor tissues selectively and provided ultrasound signals with prolonged duration and that were significantly stronger than non-targeted NBs, single-targeted NBs and SonoVue MBs. Overall, the dual targeted NBs served as efficient UCAs for specific imaging of breast cancer, and hold great potential for general cancer diagnosis/monitoring in the future.
Collapse
Affiliation(s)
- Xue Mi
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Xinmeng Guo
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Haiqiao Du
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Min Han
- Second Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Hong Liu
- Second Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Yukun Luo
- Department of Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Dekun Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Rong Xiang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Shijing Yue
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yuying Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| | - Xiaoyue Tan
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| |
Collapse
|
7
|
Gurwin A, Kowalczyk K, Knecht-Gurwin K, Stelmach P, Nowak Ł, Krajewski W, Szydełko T, Małkiewicz B. Alternatives for MRI in Prostate Cancer Diagnostics-Review of Current Ultrasound-Based Techniques. Cancers (Basel) 2022; 14:1859. [PMID: 35454767 PMCID: PMC9028694 DOI: 10.3390/cancers14081859] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
The purpose of this review is to present the current role of ultrasound-based techniques in the diagnostic pathway of prostate cancer (PCa). With overdiagnosis and overtreatment of a clinically insignificant PCa over the past years, multiparametric magnetic resonance imaging (mpMRI) started to be recommended for every patient suspected of PCa before performing a biopsy. It enabled targeted sampling of the suspicious prostate regions, improving the accuracy of the traditional systematic biopsy. However, mpMRI is associated with high costs, relatively low availability, long and separate procedure, or exposure to the contrast agent. The novel ultrasound modalities, such as shear wave elastography (SWE), contrast-enhanced ultrasound (CEUS), or high frequency micro-ultrasound (MicroUS), may be capable of maintaining the performance of mpMRI without its limitations. Moreover, the real-time lesion visualization during biopsy would significantly simplify the diagnostic process. Another value of these new techniques is the ability to enhance the performance of mpMRI by creating the image fusion of multiple modalities. Such models might be further analyzed by artificial intelligence to mark the regions of interest for investigators and help to decide about the biopsy indications. The dynamic development and promising results of new ultrasound-based techniques should encourage researchers to thoroughly study their utilization in prostate imaging.
Collapse
Affiliation(s)
- Adam Gurwin
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.K.); (P.S.); (Ł.N.); (W.K.); (T.S.)
| | - Kamil Kowalczyk
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.K.); (P.S.); (Ł.N.); (W.K.); (T.S.)
| | - Klaudia Knecht-Gurwin
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Paweł Stelmach
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.K.); (P.S.); (Ł.N.); (W.K.); (T.S.)
| | - Łukasz Nowak
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.K.); (P.S.); (Ł.N.); (W.K.); (T.S.)
| | - Wojciech Krajewski
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.K.); (P.S.); (Ł.N.); (W.K.); (T.S.)
| | - Tomasz Szydełko
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.K.); (P.S.); (Ł.N.); (W.K.); (T.S.)
| | - Bartosz Małkiewicz
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.K.); (P.S.); (Ł.N.); (W.K.); (T.S.)
| |
Collapse
|
8
|
Braga M, Leow CH, Gil JH, Teh JH, Carroll L, Long NJ, Tang MX, Aboagye EO. Investigating CXCR4 expression of tumor cells and the vascular compartment: A multimodal approach. PLoS One 2021; 16:e0260186. [PMID: 34793563 PMCID: PMC8601444 DOI: 10.1371/journal.pone.0260186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/03/2021] [Indexed: 11/19/2022] Open
Abstract
The C-X-C chemokine receptor 4 (CXCR4) is G protein-coupled receptor that upon binding to its cognate ligand, can lead to tumor progression. Several CXCR4-targeted therapies are currently under investigation, and with it comes the need for imaging agents capable of accurate depiction of CXCR4 for therapeutic stratification and monitoring. PET agents enjoy the most success, but more cost-effective and radiation-free approaches such as ultrasound (US) imaging could represent an attractive alternative. In this work, we developed a targeted microbubble (MB) for imaging of vascular CXCR4 expression in cancer. A CXCR4-targeted MB was developed through incorporation of the T140 peptide into the MB shell. Binding properties of the T140-MB and control, non-targeted MB (NT-MB) were evaluated in MDA-MB-231 cells where CXCR4 expression was knocked-down (via shRNA) through optical imaging, and in the lymphoma tumor models U2932 and SuDHL8 (high and low CXCR4 expression, respectively) by US imaging. PET imaging of [18F]MCFB, a tumor-penetrating CXCR4-targeted small molecule, was used to provide whole-tumor CXCR4 readouts. CXCR4 expression and microvessel density were performed by immunohistochemistry analysis and western blot. T140-MB were formed with similar properties to NT-MB and accumulated sensitively and specifically in cells according to their CXCR4 expression. In NOD SCID mice, T140-MB persisted longer in tumors than NT-MB, indicative of target interaction, but showed no difference between U2932 and SuDHL8. In contrast, PET imaging with [18F]MCFB showed a marked difference in tumor uptake at 40-60 min post-injection between the two tumor models (p<0.05). Ex vivo analysis revealed that the large differences in CXCR4 expression between the two models are not reflected in the vascular compartment, where the MB are restricted; in fact, microvessel density and CXCR4 expression in the vasculature was comparable between U2932 and SuDHL8 tumors. In conclusion, we successfully developed a T140-MB that can be used for imaging CXCR4 expression in the tumor vasculature.
Collapse
Affiliation(s)
- Marta Braga
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Chee Hau Leow
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London, United Kingdom
| | - Javier Hernandez Gil
- Department of Chemistry, Faculty of Natural Sciences, Imperial College London, London, United Kingdom
| | - Jin H. Teh
- Department of Chemistry, Faculty of Natural Sciences, Imperial College London, London, United Kingdom
| | - Laurence Carroll
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Nicholas J. Long
- Department of Chemistry, Faculty of Natural Sciences, Imperial College London, London, United Kingdom
| | - Meng-Xing Tang
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London, United Kingdom
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
9
|
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] [Grants] [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
|
10
|
Herbst EB, Klibanov AL, Hossack JA, Mauldin FW. Dynamic Filtering of Adherent and Non-adherent Microbubble Signals Using Singular Value Thresholding and Normalized Singular Spectrum Area Techniques. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:3240-3252. [PMID: 34376299 PMCID: PMC8691388 DOI: 10.1016/j.ultrasmedbio.2021.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 06/13/2023]
Abstract
Ultrasound molecular imaging techniques rely on the separation and identification of three types of signals: static tissue, adherent microbubbles and non-adherent microbubbles. In this study, the image filtering techniques of singular value thresholding (SVT) and normalized singular spectrum area (NSSA) were combined to isolate and identify vascular endothelial growth factor receptor 2-targeted microbubbles in a mouse hindlimb tumor model (n = 24). By use of a Verasonics Vantage 256 imaging system with an L12-5 transducer, a custom-programmed pulse inversion sequence employing synthetic aperture virtual source element imaging was used to collect contrast images of mouse tumors perfused with microbubbles. SVT was used to suppress static tissue signals by 9.6 dB while retaining adherent and non-adherent microbubble signals. NSSA was used to classify microbubble signals as adherent or non-adherent with high accuracy (receiver operating characteristic area under the curve [ROC AUC] = 0.97), matching the classification performance of differential targeted enhancement. The combined SVT + NSSA filtering method also outperformed differential targeted enhancement in differentiating MB signals from all other signals (ROC AUC = 0.89) without necessitating destruction of the contrast agent. The results from this study indicate that SVT and NSSA can be used to automatically segment and classify contrast signals. This filtering method with potential real-time capability could be used in future diagnostic settings to improve workflow and speed the clinical uptake of ultrasound molecular imaging techniques.
Collapse
Affiliation(s)
- Elizabeth B Herbst
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Alexander L Klibanov
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - John A Hossack
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - F William Mauldin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
11
|
Rabelo AC, Borghesi J, Carreira ACO, Hayashi RG, Bessa F, Barreto RDSN, da Costa RP, Cantanhede Filho AJ, Carneiro FJC, Miglino MA. Calotropis procera (Aiton) Dryand (Apocynaceae) as an anti-cancer agent against canine mammary tumor and osteosarcoma cells. Res Vet Sci 2021; 138:79-89. [PMID: 34119813 DOI: 10.1016/j.rvsc.2021.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/10/2021] [Accepted: 06/03/2021] [Indexed: 12/01/2022]
Abstract
Our goal was to evaluate phytochemical characterization and the antitumor potential of Calotropis procera. The phytochemical constitution of the crude extract (CE) revealed the presence of flavonoids, glycosides and cardenolide. The MTT assay was used to evaluate the cytotoxicity of CE, methanolic (MF) and ethyl acetate fractions (EAF) of C. procera in canine osteosarcoma cells (OST), canine mammary tumor (CMT), and canine skin fibroblasts (non-tumor cell). Doxorubicin was also used as a positive control. Results showed that CE, MF and EAF promoted a decrease in the viability of OST and CMT cells and did not alter the fibroblasts viability. C. procera also decreased the number of cells, corroborating to the decrease in proliferation and the cell cycle arrest in the G0/G1 phase. It was also evaluated the cell morphology by light and fluorescence microscopy, being demonstrated a reduction in cytoplasmic and cell rounding characteristic of programmed cell death. Moreover, flow cytometry data demonstrated that CE treatment promoted increase of caspase-3 and p53, showing that the cell death was activated in OST cells. In addition, there was a decrease in CD31, VEGF, osteopontin and TGF-β after CE treatment, suggesting that CE exerts its antitumor effect by reducing angiogenesis and tumor progression in OST cells. Moreover, CMT cells showed a reduction in PCNA after treatment with MF and CE. Analyzing the data together, C. procera, especially CE, showed an antitumor potential in both OST and CMT cells, encouraging us to continue investigating its use in cancer therapy.
Collapse
Affiliation(s)
- Ana CarolinaSilveira Rabelo
- Laboratory of Stem Cell, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508 270, Brazil.
| | - Jéssica Borghesi
- Laboratory of Stem Cell, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508 270, Brazil
| | - Ana Claudia O Carreira
- Laboratory of Stem Cell, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508 270, Brazil; Center for Cellular and Molecular Therapy (NUCEL), School of Medicine, University of São Paulo (USP), São Paulo 05360-130, Brazil
| | - Rafael Gonçalves Hayashi
- Laboratory of Stem Cell, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508 270, Brazil
| | - Fernanda Bessa
- Laboratory of Stem Cell, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508 270, Brazil
| | - Rodrigo da Silva Nunes Barreto
- Laboratory of Stem Cell, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508 270, Brazil
| | - Romário Pereira da Costa
- Department of Chemistry, Federal University of São Carlos (UFSCar), São Carlos 13560-970, Brazil
| | | | - Fernando José Costa Carneiro
- Department of Chemistry, Federal Institute of Education, Science and Technology of Maranhão, Campus São Luís, Monte Castelo, Maranhão 65030-005, Brazil
| | - Maria Angélica Miglino
- Laboratory of Stem Cell, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508 270, Brazil
| |
Collapse
|
12
|
Soletti RC, de Britto MAP, Borges HL, Machado JC. Detection of Mice Colorectal Tumors by Endoluminal Ultrasound Biomicroscopic Images and Quantification of Image Augmented Gray Values Following Injection of VEGFR-2 Targeted Contrast Agent. Acad Radiol 2021; 28:808-816. [PMID: 32067837 DOI: 10.1016/j.acra.2020.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/07/2023]
Abstract
RATIONALE AND OBJECTIVES Ultrasound biomicroscopy (UBM) is a noninvasive imaging technique that can be applied in detecting colonic tumors and, once associated with an ultrasound contrast agent (UCA), can identify the molecular expression of cancer-related biomarkers, such as the vascular endothelial growth factor receptor 2 (VEGFR-2). The present work aimed to detect colonic tumors and quantify augmented gray values of endoluminal UBM (eUBM) images from colonic tumors following the injection of VEGFR-2 targeted UCA (VEGFR2-UCA) into a mouse model of colorectal cancer. MATERIAL AND METHODS A 40 MHz miniprobe catheter inserted through the biopsy channel of a pediatric flexible bronchofiberscope was used to obtain colonoscopic and B-mode eUBM images simultaneously. Seventeen tumor-bearing mice had their colons inspected and six of them were subjected to a VEGFR2-UCA injection to predict VEGFR-2 expression. RESULTS All animals developed distal colon tumors and eUBM was able to detect all of them and also to characterize the tumors, with 71.4% being in situ lesions and 28.6% being tumors invading the mucosa + muscularis mucosae + submucosa layers, as confirmed by histopathology. After VEGFR2-UCA injection, gray values from the eUBM tumoral images increased significantly (p < 0.01). Tumor sites with increased eUBM image gray values corresponded to areas with increased VEGFR-2 expression, as confirmed by immunohistochemistry. CONCLUSION The results confirm eUBM as a powerful noninvasive and real-time tool for detecting colon tumor and its invasiveness and once associated with VEGFR2-UCA may become a tool for the detection of VEGFR-2 expression in colonic tumors.
Collapse
|
13
|
Sier VQ, van der Vorst JR, Quax PHA, de Vries MR, Zonoobi E, Vahrmeijer AL, Dekkers IA, de Geus-Oei LF, Smits AM, Cai W, Sier CFM, Goumans MJTH, Hawinkels LJAC. Endoglin/CD105-Based Imaging of Cancer and Cardiovascular Diseases: A Systematic Review. Int J Mol Sci 2021; 22:4804. [PMID: 33946583 PMCID: PMC8124553 DOI: 10.3390/ijms22094804] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Molecular imaging of pathologic lesions can improve efficient detection of cancer and cardiovascular diseases. A shared pathophysiological feature is angiogenesis, the formation of new blood vessels. Endoglin (CD105) is a coreceptor for ligands of the Transforming Growth Factor-β (TGF-β) family and is highly expressed on angiogenic endothelial cells. Therefore, endoglin-based imaging has been explored to visualize lesions of the aforementioned diseases. This systematic review highlights the progress in endoglin-based imaging of cancer, atherosclerosis, myocardial infarction, and aortic aneurysm, focusing on positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), near-infrared fluorescence (NIRF) imaging, and ultrasound imaging. PubMed was searched combining the following subjects and their respective synonyms or relevant subterms: "Endoglin", "Imaging/Image-guided surgery". In total, 59 papers were found eligible to be included: 58 reporting about preclinical animal or in vitro models and one ex vivo study in human organs. In addition to exact data extraction of imaging modality type, tumor or cardiovascular disease model, and tracer (class), outcomes were described via a narrative synthesis. Collectively, the data identify endoglin as a suitable target for intraoperative and diagnostic imaging of the neovasculature in tumors, whereas for cardiovascular diseases, the evidence remains scarce but promising.
Collapse
Affiliation(s)
- Vincent Q. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Joost R. van der Vorst
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Paul H. A. Quax
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Margreet R. de Vries
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Elham Zonoobi
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
- Edinburgh Molecular Imaging Ltd. (EMI), Edinburgh EH16 4UX, UK
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Ilona A. Dekkers
- Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7500 AE Enschede, The Netherlands
| | - Anke M. Smits
- Department of Cell & Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (A.M.S.); (M.J.T.H.G.)
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Marie José T. H. Goumans
- Department of Cell & Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (A.M.S.); (M.J.T.H.G.)
| | - Lukas J. A. C. Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| |
Collapse
|
14
|
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
|
15
|
Kis A, Dénes N, Szabó JP, Arató V, Jószai I, Enyedi KN, Lakatos S, Garai I, Mező G, Kertész I, Trencsényi G. In vivo assessment of aminopeptidase N (APN/CD13) specificity of different 68Ga-labelled NGR derivatives using PET/MRI imaging. Int J Pharm 2020; 589:119881. [PMID: 32946975 DOI: 10.1016/j.ijpharm.2020.119881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Aminopeptidase N (APN/CD13) plays an important role in neoangiogenic process in malignancies. Our previous studies have already shown that 68Ga-labelled NOTA conjugated asparagine-glycine-arginine peptide (c[KNGRE]-NH2) specifically bind to APN/CD13 expressing tumors. The aim of this study was to evaluate and compare the APN/CD13 specificity of newly synthesized 68Ga-labelled NGR derivatives in vivo by PET/MRI imaging using hepatocellular carcinoma (He/De) and mesoblastic nephroma (Ne/De) tumor models. PET/MRI and ex vivo biodistribution studies were performed 11 ± 1 days after subcutaneous injection of tumor cells and 90 min after intravenous injection of 68Ga-NOTA-c(NGR), 68Ga-NODAGA-c(NGR), 68Ga-NODAGA-c(NGR) (MG1) or 68Ga-NODAGA-c(NGR) (MG2). The APN/CD13 selectivity was confirmed by blocking experiments and the APN/CD13 expression was verified by immunohistochemistry. 68Ga-labelled c(NGR) derivatives were produced with high specific activity and radiochemical purity. In control animals, low radiotracer accumulation was found in abdominal and thoracic organs. Using tumor-bearing animals we found that the 68Ga-NOTA-c(NGR), 68Ga-NODAGA-c(NGR), and 68Ga-NODAGA-c(NGR) (MG1) derivatives showed higher uptake in He/De and Ne/De tumors, than that of the accumulation of 68Ga-NODAGA-c(NGR) (MG2). APN/CD13 is a very promising target in PET imaging, however, the selection of the appropriate 68Ga-labelled NGR-based radiopharmaceutical is critical for the precise detection of tumor neo-angiogenesis and for monitoring the efficacy of anticancer therapy.
Collapse
Key Words
- (68)Ga
- Aminopeptidase N
- Angiogenesis
- CD13
- CID: 2796029, 1-hydroxybenzotriazole (HOBt)
- CID: 3036142, 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA)
- CID: 33032, L-Glutamic acid
- CID: 5962, L-Lysine
- CID: 6228, N,N-dimethylformamide (DMF)
- CID: 6267, L-Asparagine
- CID: 6322, L-Arginine
- CID: 6422, triflouroacetic acid (TFA)
- CID: 750, Glyicine
- NGR
- PET/MRI imaging
Collapse
Affiliation(s)
- Adrienn Kis
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary; Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Noémi Dénes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary; Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Judit P Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary; Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Viktória Arató
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary
| | - István Jószai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary
| | - Kata Nóra Enyedi
- Eötvös Loránd University, Faculty of Science, Institute of Chemistry, Budapest, Hungary
| | - Szilvia Lakatos
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary
| | - Ildikó Garai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary; Scanomed LTD, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Gábor Mező
- Eötvös Loránd University, Faculty of Science, Institute of Chemistry, Budapest, Hungary; MTA-ELTE, Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös L. University, Budapest, Hungary
| | - István Kertész
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Hungary; Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary; Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary.
| |
Collapse
|
16
|
Nie Z, Luo N, Liu J, Zhang Y, Zeng X, Su D. Dual-Mode Contrast Agents with RGD-Modified Polymer for Tumour-Targeted US/NIRF Imaging. Onco Targets Ther 2020; 13:8919-8929. [PMID: 32982284 PMCID: PMC7495348 DOI: 10.2147/ott.s256044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/06/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Cancer diagnosis and treatment during the early stages of disease remain extremely challenging clinical tasks. The development of effective multimode contrast agents could greatly facilitate the early detection of cancer. MATERIALS AND METHODS We prepared dual-mode contrast agents using a biotin/avidin bioamplification system. Through in vivo and in vitro experiments, we verified the imaging performance of this contrast agents in both fluorescence and ultrasound and its targeting specificity for MDA-MB-231 cells. RESULTS The RGD peptide-labelled microbubbles showed excellent targeting of αvβ3 integrin expressed by MDA-MB-231 cells in vitro and in vivo. The signal intensity and time duration of ultrasound imaging using these particles were superior to those obtained with a typical ultrasound contrast agent in the clinic. The tumour areas also demonstrated high Cy5.5 accumulation by fluorescence imaging. CONCLUSION The results show that this targeted dual-mode imaging system yields outstanding US/NIRF imaging results, possibly allowing the early clinical diagnosis of cancer.
Collapse
Affiliation(s)
- Zhenhui Nie
- Department of Radiology, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Ningbin Luo
- Department of Radiology, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Junjie Liu
- Department of Medical Ultrasound, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Yu Zhang
- Department of Radiology, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Xinyi Zeng
- Department of Radiology, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Danke Su
- Department of Radiology, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| |
Collapse
|
17
|
Ingram N, McVeigh LE, Abou-Saleh RH, Maynard J, Peyman SA, McLaughlan JR, Fairclough M, Marston G, Valleley EMA, Jimenez-Macias JL, Charalambous A, Townley W, Haddrick M, Wierzbicki A, Wright A, Volpato M, Simpson PB, Treanor DE, Thomson NH, Loadman PM, Bushby RJ, Johnson BR, Jones PF, Evans JA, Freear S, Markham AF, Evans SD, Coletta PL. Ultrasound-triggered therapeutic microbubbles enhance the efficacy of cytotoxic drugs by increasing circulation and tumor drug accumulation and limiting bioavailability and toxicity in normal tissues. Theranostics 2020; 10:10973-10992. [PMID: 33042265 PMCID: PMC7532679 DOI: 10.7150/thno.49670] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Most cancer patients receive chemotherapy at some stage of their treatment which makes improving the efficacy of cytotoxic drugs an ongoing and important goal. Despite large numbers of potent anti-cancer agents being developed, a major obstacle to clinical translation remains the inability to deliver therapeutic doses to a tumor without causing intolerable side effects. To address this problem, there has been intense interest in nanoformulations and targeted delivery to improve cancer outcomes. The aim of this work was to demonstrate how vascular endothelial growth factor receptor 2 (VEGFR2)-targeted, ultrasound-triggered delivery with therapeutic microbubbles (thMBs) could improve the therapeutic range of cytotoxic drugs. Methods: Using a microfluidic microbubble production platform, we generated thMBs comprising VEGFR2-targeted microbubbles with attached liposomal payloads for localised ultrasound-triggered delivery of irinotecan and SN38 in mouse models of colorectal cancer. Intravenous injection into tumor-bearing mice was used to examine targeting efficiency and tumor pharmacodynamics. High-frequency ultrasound and bioluminescent imaging were used to visualise microbubbles in real-time. Tandem mass spectrometry (LC-MS/MS) was used to quantitate intratumoral drug delivery and tissue biodistribution. Finally, 89Zr PET radiotracing was used to compare biodistribution and tumor accumulation of ultrasound-triggered SN38 thMBs with VEGFR2-targeted SN38 liposomes alone. Results: ThMBs specifically bound VEGFR2 in vitro and significantly improved tumor responses to low dose irinotecan and SN38 in human colorectal cancer xenografts. An ultrasound trigger was essential to achieve the selective effects of thMBs as without it, thMBs failed to extend intratumoral drug delivery or demonstrate enhanced tumor responses. Sensitive LC-MS/MS quantification of drugs and their metabolites demonstrated that thMBs extended drug exposure in tumors but limited exposure in healthy tissues, not exposed to ultrasound, by persistent encapsulation of drug prior to elimination. 89Zr PET radiotracing showed that the percentage injected dose in tumors achieved with thMBs was twice that of VEGFR2-targeted SN38 liposomes alone. Conclusions: thMBs provide a generic platform for the targeted, ultrasound-triggered delivery of cytotoxic drugs by enhancing tumor responses to low dose drug delivery via combined effects on circulation, tumor drug accumulation and exposure and altered metabolism in normal tissues.
Collapse
Affiliation(s)
- Nicola Ingram
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Laura E. McVeigh
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Radwa H. Abou-Saleh
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
- Department of Physics, Faculty of Science, Mansoura University, Egypt
| | - Juliana Maynard
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Sally A. Peyman
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - James R. McLaughlan
- Faculty of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Michael Fairclough
- Wolfson Molecular Imaging Centre, University of Manchester, Palatine Road, Manchester, M20 3LI, United Kingdom
| | - Gemma Marston
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Elizabeth M. A. Valleley
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Jorge L. Jimenez-Macias
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Antonia Charalambous
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - William Townley
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Antonia Wierzbicki
- Institute of Cancer Therapeutics, University of Bradford, BD7 1DP, United Kingdom
| | - Alexander Wright
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Milène Volpato
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Peter B. Simpson
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Darren E. Treanor
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Neil H. Thomson
- School of Dentistry, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Paul M. Loadman
- Institute of Cancer Therapeutics, University of Bradford, BD7 1DP, United Kingdom
| | - Richard J. Bushby
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Benjamin R.G. Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - Pamela F. Jones
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - J. Anthony Evans
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Steven Freear
- Faculty of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Alexander F. Markham
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Stephen D. Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - P. Louise Coletta
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| |
Collapse
|
18
|
In Vivo Imaging of Hypoxia and Neoangiogenesis in Experimental Syngeneic Hepatocellular Carcinoma Tumor Model Using Positron Emission Tomography. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4952372. [PMID: 32832549 PMCID: PMC7428931 DOI: 10.1155/2020/4952372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/29/2020] [Accepted: 07/11/2020] [Indexed: 12/19/2022]
Abstract
Introduction Hypoxia-induced ανβ3 integrin and aminopeptidase N (APN/CD13) receptor expression play an important role in tumor neoangiogenesis. APN/CD13-specific 68Ga-NOTA-c(NGR), ανβ3 integrin-specific 68Ga-NODAGA-[c(RGD)]2, and hypoxia-specific 68Ga-DOTA-nitroimidazole enable the in vivo detection of the neoangiogenic process and the hypoxic regions in the tumor mass using positron emission tomography (PET) imaging. The aim of this study was to evaluate whether 68Ga-NOTA-c(NGR) and 68Ga-DOTA-nitroimidazole allow the in vivo noninvasive detection of the temporal changes of APN/CD13 expression and hypoxia in experimental He/De tumors using positron emission tomography. Materials and Methods 5 × 106 hepatocellular carcinoma (He/De) cells were used for the induction of a subcutaneous tumor model in Fischer-344 rats. He/De tumor-bearing animals were anaesthetized, and 90 min after intravenous injection of 10.2 ± 1.1 MBq 68Ga-NOTA-c(NGR) or 68Ga-NODAGA-[c(RGD)]2 (as angiogenesis tracers) or 68Ga-DOTA-nitroimidazole (for hypoxia imaging), whole-body PET/MRI scans were performed. Results Hypoxic regions and angiogenic markers (αvβ3 integrin and APN/CD13) were determined using 68Ga-NOTA-c(NGR), 68Ga-DOTA-nitroimidazole, and 68Ga-NODAGA-[c(RGD)]2 in subcutaneously growing He/De tumors in rats. 68Ga-NOTA-c(NGR) showed the strong APN/CD13 positivity of He/De tumors in vivo, by which observation was confirmed by western blot analysis. By the qualitative analysis of PET images, heterogenous accumulation was found inside He/De tumors using all radiotracers. Significantly (p ≤ 0.01) higher SUVmean and SUVmax values were found in the radiotracer avid regions of the tumors than those of the nonavid areas using hypoxia and angiogenesis-specific radiopharmaceuticals. Furthermore, a strong correlation was found between the presence of angiogenic markers, the appearance of hypoxic regions, and the tumor volume using noninvasive in vivo PET imaging. Conclusion 68Ga-DOTA-nitroimidazole and 68Ga-NOTA-c(NGR) are suitable diagnostic radiotracers for the detection of the temporal changes of hypoxic areas and neoangiogenic molecule (CD13) expression, which vary during tumor growth in a hepatocellular carcinoma model.
Collapse
|
19
|
Kosareva A, Abou-Elkacem L, Chowdhury S, Lindner JR, Kaufmann BA. Seeing the Invisible-Ultrasound Molecular Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:479-497. [PMID: 31899040 DOI: 10.1016/j.ultrasmedbio.2019.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 06/10/2023]
Abstract
Ultrasound molecular imaging has been developed in the past two decades with the goal of non-invasively imaging disease phenotypes on a cellular level not depicted on anatomic imaging. Such techniques already play a role in pre-clinical research for the assessment of disease mechanisms and drug effects, and are thought to in the future contribute to earlier diagnosis of disease, assessment of therapeutic effects and patient-tailored therapy in the clinical field. In this review, we first describe the chemical composition and structure as well as the in vivo behavior of the ultrasound contrast agents that have been developed for molecular imaging. We then discuss the strategies that are used for targeting of contrast agents to specific cellular targets and protocols used for imaging. Next we describe pre-clinical data on imaging of thrombosis, atherosclerosis and microvascular inflammation and in oncology, including the pathophysiological principles underlying the selection of targets in each area. Where applicable, we also discuss efforts that are currently underway for translation of this technique into the clinical arena.
Collapse
Affiliation(s)
- Alexandra Kosareva
- Cardiovascular Molecular Imaging, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Lotfi Abou-Elkacem
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California, USA
| | - Sayan Chowdhury
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Portland, Oregon, USA; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Beat A Kaufmann
- Cardiovascular Molecular Imaging, Department of Biomedicine, University of Basel, Basel, Switzerland; Department of Cardiology, University Hospital and University of Basel, Basel, Switzerland.
| |
Collapse
|
20
|
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
|
21
|
Lau C, Rivas M, Dinalo J, King K, Duddalwar V. Scoping Review of Targeted Ultrasound Contrast Agents in the Detection of Angiogenesis. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2020; 39:19-28. [PMID: 31237009 DOI: 10.1002/jum.15072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 06/09/2023]
Abstract
A systematic search was conducted to categorize targeted ultrasound contrast agents (UCAs) used in cancer-related angiogenesis detection. We identified 15 unique contrast agents from 2008 to March 2018. Most primary research articles studied UCAs targeted to vascular endothelial growth factor receptor or αv β3 -integrin. Breast cancer and colon cancer are the most common neoplastic processes in which these agents were studied. BR55 (Bracco Research SA, Geneva, Switzerland), a vascular endothelial growth factor receptor-targeting UCA, is the first targeted UCA that has completed phase 0 trials. Our review identifies a gap in the literature regarding the application of targeted UCAs in cancer models beyond breast and colon cancers and identifies other promising UCAs.
Collapse
Affiliation(s)
- Christopher Lau
- Department of Radiology, Keck School of Medicine, California, Los Angeles, USA
| | - Marielena Rivas
- Department of Radiology, Keck School of Medicine, California, Los Angeles, USA
| | - Jennifer Dinalo
- Norris Medical Library, Keck School of Medicine, California, Los Angeles, USA
| | - Kevin King
- Department of Radiology, Keck School of Medicine, California, Los Angeles, USA
| | - Vinay Duddalwar
- Department of Radiology, Keck School of Medicine, California, Los Angeles, USA
| |
Collapse
|
22
|
Preclinical Molecular Imaging for Precision Medicine in Breast Cancer Mouse Models. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:8946729. [PMID: 31598114 PMCID: PMC6778915 DOI: 10.1155/2019/8946729] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/28/2019] [Accepted: 07/25/2019] [Indexed: 12/18/2022]
Abstract
Precision and personalized medicine is gaining importance in modern clinical medicine, as it aims to improve diagnostic precision and to reduce consequent therapeutic failures. In this regard, prior to use in human trials, animal models can help evaluate novel imaging approaches and therapeutic strategies and can help discover new biomarkers. Breast cancer is the most common malignancy in women worldwide, accounting for 25% of cases of all cancers and is responsible for approximately 500,000 deaths per year. Thus, it is important to identify accurate biomarkers for precise stratification of affected patients and for early detection of responsiveness to the selected therapeutic protocol. This review aims to summarize the latest advancements in preclinical molecular imaging in breast cancer mouse models. Positron emission tomography (PET) imaging remains one of the most common preclinical techniques used to evaluate biomarker expression in vivo, whereas magnetic resonance imaging (MRI), particularly diffusion-weighted (DW) sequences, has been demonstrated as capable of distinguishing responders from nonresponders for both conventional and innovative chemo- and immune-therapies with high sensitivity and in a noninvasive manner. The ability to customize therapies is desirable, as this will enable early detection of diseases and tailoring of treatments to individual patient profiles. Animal models remain irreplaceable in the effort to understand the molecular mechanisms and patterns of oncologic diseases.
Collapse
|
23
|
Herbst EB, Unnikrishnan S, Klibanov AL, Mauldin FW, Hossack JA. Validation of Normalized Singular Spectrum Area as a Classifier for Molecularly Targeted Microbubble Adherence. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2493-2501. [PMID: 31227262 PMCID: PMC7480935 DOI: 10.1016/j.ultrasmedbio.2019.05.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 05/24/2023]
Abstract
Ultrasound molecular imaging is a diagnostic technique wherein molecularly targeted microbubble contrast agents are imaged to reveal disease markers on the blood vessel endothelium. Currently, microbubble adhesion to affected tissue can be quantified using differential targeted enhancement (dTE), which measures the late enhancement of adherent microbubbles through administration of destructive ultrasound pressures. In this study, we investigated a statistical parameter called the normalized singular spectrum area (NSSA) as a means to detect microbubble adhesion without microbubble destruction. We compared the signal differentiation capability of NSSA with matched dTE measurements in a mouse hindlimb tumor model. Results indicated that NSSA-based signal classification performance matches dTE when differentiating adherent microbubble from non-adherent microbubble signals (receiver operating characteristic area under the curve = 0.95), and improves classification performance when differentiating microbubble from tissue signals (p < 0.005). NSSA-based signal classification eliminates the need for destruction of contrast, and may offer better sensitivity, specificity and the opportunity for real-time microbubble detection and classification.
Collapse
Affiliation(s)
- Elizabeth B Herbst
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Sunil Unnikrishnan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Alexander L Klibanov
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - F William Mauldin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - John A Hossack
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
24
|
Wu H, Abenojar EC, Perera R, De Leon AC, An T, Exner AA. Time-intensity-curve Analysis and Tumor Extravasation of Nanobubble Ultrasound Contrast Agents. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2502-2514. [PMID: 31248638 PMCID: PMC6689247 DOI: 10.1016/j.ultrasmedbio.2019.05.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/28/2019] [Accepted: 05/22/2019] [Indexed: 05/05/2023]
Abstract
Our group recently presented a simple strategy using the non-ionic surfactant, Pluronic, as a size control excipient to produce nanobubbles in the 100-nm range, which exhibited stability and echogenicity on par with clinically available microbubbles. The objective of the present study was to evaluate biodistribution and extravasation of the Pluronic-stabilized lipid nanobubbles compared with microbubbles in 2 experimental tumor models in mice. Standard lipid-stabilized perfluoropropane bubbles (Pluronic L10) and lipid-stabilized perfluoropropane nanobubbles were intravenously injected into mice bearing either an orthotopic mouse breast cancer (BC4 T1) or subcutaneous mouse ovarian cancer (OVCAR-3) through the tail vein to perform perfusion dynamic studies. No significant differences between the nanobubble and microbubble groups were observed in the peak enhancement of the 3 tested regions (tumor, liver and kidney). However, the decay rates of nanobubble in the tumor and kidney of BC4 T1-bearing mice, as well as in mice with OVRCAR-3 tumors were significantly slower than those of the microbubble. To quantify extravasation, fluorescently labeled bubbles were intravenously injected into mice bearing the same tumors. Histologic analysis showed that nanobubbles were retained in tumor tissue to a greater extent compared with microbubbles in both tumor models at the 3-h time point. Our results demonstrate unique nanobubble behavior compared with microbubbles and support augmented application of these agents in ultrasound molecular imaging and drug delivery beyond the tumor vasculature.
Collapse
Affiliation(s)
- Hanping Wu
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Eric C Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Reshani Perera
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | | | - Tianzhi An
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
25
|
Jing Y, Xiu-Juan Z, Hong-Jiao C, Zhi-Kui C, Qing-Fu Q, En-Sheng X, Li-Wu L. Ultrasound-targeted microbubble destruction improved the antiangiogenic effect of Endostar in triple-negative breast carcinoma xenografts. J Cancer Res Clin Oncol 2019; 145:1191-1200. [DOI: 10.1007/s00432-019-02866-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 02/16/2019] [Indexed: 12/15/2022]
|
26
|
Nam K, Stanczak M, Forsberg F, Liu JB, Eisenbrey JR, Solomides CC, Lyshchik A. Sentinel Lymph Node Characterization with a Dual-Targeted Molecular Ultrasound Contrast Agent. Mol Imaging Biol 2019; 20:221-229. [PMID: 28762204 DOI: 10.1007/s11307-017-1109-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The purpose of this study was to assess the performance of molecular ultrasound with dual-targeted microbubbles to detect metastatic disease in the sentinel lymph nodes (SLNs) in swine model of naturally occurring melanoma. The SLN is the first lymph node in the lymphatic chain draining primary tumor, and early detection of metastatic SLN involvement is critical in the appropriate management of melanoma. PROCEDURE Nine Sinclair swine (weight 3-7 kg; Sinclair BioResources, Columbia, MO, USA) with naturally occurring melanoma were examined. Siemens S3000 scanner with a 9L4 probe was used for imaging (Siemens Healthineers, Mountain View, CA). Dual-targeted contrast agent was created using Targestar SA microbubbles (Targeson, San Diego, CA, USA) labeled with ανβ3-integrin and P-selectin antibodies. Targestar SA microbubbles labeled with IgG-labeled were used as control. First, peritumoral injection of Sonazoid contrast agent (GE Healthcare, Oslo, Norway) was performed to detect SLNs. After that, dual-targeted and IGG control Targestar SA microbubbles were injected intravenously with a 30-min interval between injections. Labeled Targestar SA microbubbles were allowed to circulate for 4 min to enable binding. After that, two sets of image clips were acquired several seconds before and after a high-power destruction sequence. The mean intensity difference pre- to post-bubble destruction within the region of interest placed over SLN was calculated as a relative measure of targeted microbubble contrast agent retention. This process was repeated for non-SLNs as controls. All lymph nodes evaluated on imaging were surgically removed and histologically examined for presence of metastatic involvement. RESULTS A total of 43 lymph nodes (25 SLNs and 18 non-SLNs) were included in the analysis with 18 SLNs demonstrating metastatic involvement greater than 5 % on histology. All non-SLNs were benign. The mean intensity (± SD) of the dual-targeted microbubbles for metastatic SLNs was significantly higher than that of benign LNs (18.05 ± 19.11 vs. 3.30 ± 6.65 AU; p = 0.0008), while IgG-labeled control microbubbles demonstrated no difference in retained contrast intensity between metastatic and benign lymph nodes (0.39 ± 1.14 vs. 0.03 ± 0.24 AU; p = 0.14). CONCLUSIONS The results indicate that dual-targeted microbubbles labeled with P-selectin and ανβ3-integrin antibodies may aid in detecting metastatic involvement in SLNs of melanoma.
Collapse
Affiliation(s)
- Kibo Nam
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, Philadelphia, PA, 19107, USA
| | - Maria Stanczak
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, Philadelphia, PA, 19107, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, Philadelphia, PA, 19107, USA
| | - Ji-Bin Liu
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, Philadelphia, PA, 19107, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, Philadelphia, PA, 19107, USA
| | | | - Andrej Lyshchik
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, Philadelphia, PA, 19107, USA.
| |
Collapse
|
27
|
InVivo Molecular Ultrasound Assessment of Glioblastoma Neovasculature with Endoglin-Targeted Microbubbles. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:8425495. [PMID: 30498402 PMCID: PMC6220748 DOI: 10.1155/2018/8425495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/16/2018] [Accepted: 09/20/2018] [Indexed: 12/16/2022]
Abstract
Objectives Glioblastoma, as one of the most malignant cancer in the world, usually shows substantially increased angiogenesis. Endoglin (CD105), which is an alternative proangiogenic growth factor, has been remarkably upregulated on the proliferating glioblastoma neovasculature. However, little is known on the noninvasive assessment of the expression levels of CD105 during glioblastoma progression. Herein, we investigated the potential of the molecular ultrasound imaging for the noninvasive assessment of the expression levels of the biomarker CD105 during the glioblastoma progression. Materials and Methods The CD105-targeted perfluorocarbon-containing lipid-shelled microbubbles (MBs) were prepared. A parallel flow chamber was employed, in which the CD105-targeted and non-targeted MBs were tested across the CD105 ± expression cell lines. In vivo molecular US imaging was conducted based on a subcutaneous xenograft tumor model (n=9). Finally, the statistical analysis was conducted to quantitatively correlate the attachment numbers of MBs in the parallel flow chamber test with the CD105 expression levels of the cells in the flow cytometry test and the in vivo molecular ultrasound signals with the ex vivo expression levels of CD105 in the immunohistochemical test. Results and Discussion The attachment numbers of the CD105-targeted MBs significantly correlated with the CD105 expression levels of the cells in the parallel flow chamber test. There was a good correlation between the in vivo molecular ultrasound signals with the CD105-targeted MBs and the ex vivo expression levels of CD105 in the immunohistochemical test. The results indicate that the molecular US imaging is much potential to assess the progression of the glioblastoma neovasculature noninvasively.
Collapse
|
28
|
Wischhusen J, Wilson KE, Delcros JG, Molina-Peña R, Gibert B, Jiang S, Ngo J, Goldschneider D, Mehlen P, Willmann JK, Padilla F. Ultrasound molecular imaging as a non-invasive companion diagnostic for netrin-1 interference therapy in breast cancer. Theranostics 2018; 8:5126-5142. [PMID: 30429890 PMCID: PMC6217066 DOI: 10.7150/thno.27221] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023] Open
Abstract
In ultrasound molecular imaging (USMI), ligand-functionalized microbubbles (MBs) are used to visualize vascular endothelial targets. Netrin-1 is upregulated in 60% of metastatic breast cancers and promotes tumor progression. A novel netrin-1 interference therapy requires the assessment of netrin-1 expression prior to treatment. In this study, we studied netrin-1 as a target for USMI and its potential as a companion diagnostic in breast cancer models. Methods: To verify netrin-1 expression and localization, an in vivo immuno-localization approach was applied, in which anti-netrin-1 antibody was injected into living mice 24 h before tumor collection, and revealed with secondary fluorescent antibody for immunofluorescence analysis. Netrin-1 interactions with the cell surface were studied by flow cytometry. Netrin-1-targeted MBs were prepared using MicroMarker Target-Ready (VisualSonics), and validated in in vitro binding assays in static conditions or in a flow chamber using purified netrin-1 protein or netrin-1-expressing cancer cells. In vivo USMI of netrin-1 was validated in nude mice bearing human netrin-1-positive SKBR7 tumors or weakly netrin-1-expressing MDA-MB-231 tumors using the Vevo 2100 small animal imaging device (VisualSonics). USMI feasibility was further tested in transgenic murine FVB/N Tg(MMTV/PyMT634Mul) (MMTV-PyMT) mammary tumors. Results: Netrin-1 co-localized with endothelial CD31 in netrin-1-positive breast tumors. Netrin-1 binding to the surface of endothelial HUVEC and cancer cells was partially mediated by heparan sulfate proteoglycans. MBs targeted with humanized monoclonal anti-netrin-1 antibody bound to netrin-1-expressing cancer cells in static and dynamic conditions. USMI signal was significantly increased with anti-netrin-1 MBs in human SKBR7 breast tumors and transgenic murine MMTV-PyMT mammary tumors compared to signals recorded with either isotype control MBs or after blocking of netrin-1 with humanized monoclonal anti-netrin-1 antibody. In weakly netrin-1-expressing human tumors and normal mammary glands, no difference in imaging signal was observed with anti-netrin-1- and isotype control MBs. Ex vivo analysis confirmed netrin-1 expression in MMTV-PyMT tumors. Conclusions: These results show that USMI allowed reliable detection of netrin-1 on the endothelium of netrin-1-positive human and murine tumors. Significant differences in USMI signal for netrin-1 reflected the significant differences in netrin-1 mRNA & protein expression observed between different breast tumor models. The imaging approach was non-invasive and safe, and provided the netrin-1 expression status in near real-time. Thus, USMI of netrin-1 has the potential to become a companion diagnostic for the stratification of patients for netrin-1 interference therapy in future clinical trials.
Collapse
|
29
|
Shan R, Wang B, Wang A, Sun Z, Dong F, Liu J, Sun H. Endoglin-targeted contrast-enhanced ultrasound imaging in hepatoblastoma xenografts. Oncol Lett 2018; 16:3784-3790. [PMID: 30127989 PMCID: PMC6096263 DOI: 10.3892/ol.2018.9067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 06/04/2018] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is required for the growth of hepatoblastoma (HB). In the present study, an ultrasonic contrast agent, microbubbles (MB), was combined with an endoglin antibody, and then injected into nude mice with HB. This was conducted to detect specific binding to microvessels via non-linear harmonic imaging for tumor angiogenesis assessment. In addition, endoglin expression in experimental animals was measured using western blotting, reverse transcription-quantitative polymerase chain reaction and immunohistochemistry. In vitro, human umbilical vein endothelial cells (HUVECs) were co-cultured with conditioned media collected from HepG2 cells. Western blotting and reverse transcription-quantitative PCR was performed to detect the changes of endoglin expression. In targeted ultrasound imaging, it was determined that the differential targeted enhancement of MBendoglin was significantly higher than that of MBisotype. Over expression of endoglin was identified in the tumor of experimental nude mice; however, it was not present in the liver of the mice. Endoglin expression in HUVECs was significantly increased by co-culture with the conditioned media of HepG2 cells; therefore, the results suggest that endoglin is upregulated in angiogenic vessels in the HepG2 cell xenografts in nude mice. Thus, endoglin-targeted ultrasound imaging is presented as a potential approach for the diagnosis of liver carcinoma.
Collapse
Affiliation(s)
- Rong Shan
- Department of Ultrasonography, Jinan Infectious Disease Hospital, Jinan, Shandong 250021, P.R. China.,Department of Ultrasound, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Bei Wang
- Department of Ultrasonography, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Aiguang Wang
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Zongguo Sun
- The Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Fengyun Dong
- The Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Ju Liu
- The Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Hongjun Sun
- Department of Ultrasonography, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
30
|
Tummers WS, Willmann JK, Bonsing BA, Vahrmeijer AL, Gambhir SS, Swijnenburg RJ. Advances in Diagnostic and Intraoperative Molecular Imaging of Pancreatic Cancer. Pancreas 2018; 47:675-689. [PMID: 29894417 PMCID: PMC6003672 DOI: 10.1097/mpa.0000000000001075] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis. To improve outcomes, there is a critical need for improved tools for detection, accurate staging, and resectability assessment. This could improve patient stratification for the most optimal primary treatment modality. Molecular imaging, used in combination with tumor-specific imaging agents, can improve established imaging methods for PDAC. These novel, tumor-specific imaging agents developed to target specific biomarkers have the potential to specifically differentiate between malignant and benign diseases, such as pancreatitis. When these agents are coupled to various types of labels, this type of molecular imaging can provide integrated diagnostic, noninvasive imaging of PDAC as well as image-guided pancreatic surgery. This review provides a detailed overview of the current clinical imaging applications, upcoming molecular imaging strategies for PDAC, and potential targets for imaging, with an emphasis on intraoperative imaging applications.
Collapse
Affiliation(s)
- Willemieke S. Tummers
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA. Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Juergen K. Willmann
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA. Juergen K. Willmann died January 8, 2018
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sanjiv S. Gambhir
- Address correspondence to: R.J. Swijnenburg, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands (). Tel: +31 71 526 4005, Fax: +31 71 526 6750
| | - Rutger-Jan Swijnenburg
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
31
|
Yan H, Gao X, Zhang Y, Chang W, Li J, Li X, Du Q, Li C. Imaging Tiny Hepatic Tumor Xenografts via Endoglin-Targeted Paramagnetic/Optical Nanoprobe. ACS APPLIED MATERIALS & INTERFACES 2018; 10:17047-17057. [PMID: 29708329 DOI: 10.1021/acsami.8b02648] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Surgery is the mainstay for treating hepatocellular carcinoma (HCC). However, it is a great challenge for surgeons to identify HCC in its early developmental stage. The diagnostic sensitivity for a tiny HCC with a diameter less than 1.0 cm is usually as low as 10-33% for computed tomography (CT) and 29-43% for magnetic resonance imaging (MRI). Although MRI is the preferred imaging modality for detecting HCC, with its unparalleled spatial resolution for soft tissue, the commercially available contrast agent, such as Gd3+-DTPA, cannot accurately define HCC because of its short circulation lifetime and lack of tumor-targeting specificity. Endoglin (CD105), a type I membrane glycoprotein, is highly expressed both in HCC cells and in the endothelial cells of neovasculature, which are abundant at the tumor periphery. In this work, a novel single-stranded DNA oligonucleotide-based aptamer was screened by systematic evolution of ligands in an exponential enrichment assay and showed a high binding affinity ( KD = 98 pmol/L) to endoglin. Conjugating the aptamers and imaging reporters on a G5 dendrimer created an HCC-targeting nanoprobe that allowed the successful visualization of orthotopic HCC xenografts with diameters as small as 1-4 mm. Significantly, the invasive tumor margin was clearly delineated, with a tumor to normal ratio of 2.7 by near-infrared (NIR) fluorescence imaging and 2.1 by T1-weighted MRI. This multimodal nanoprobe holds promise not only for noninvasively defining tiny HCC by preoperative MRI but also for guiding tumor excision via intraoperative NIR fluorescence imaging, which will probably gain benefit for the patient's therapeutic response and improve the survival rate.
Collapse
Affiliation(s)
- Huihui Yan
- Department of Gastroenterology, The Second Affiliated Hospital, College of Medicine , Zhejiang University , Hangzhou , Zhejiang Province 310009 , China
| | - Xihui Gao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy , Fudan University , Shanghai 201203 , China
- Department of Laboratory Medicine & Central Laboratory , Shanghai Jiaotong University Affiliated Sixth People's Hospital South Campus , Shanghai 201499 , China
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites , Shanghai Jiao Tong University , Shanghai 200240 , China
| | - Yunfei Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Wenju Chang
- Department of General Surgery, Zhongshan Hospital , Fudan University , Shanghai 200032 , China
| | - Jianhui Li
- Ningbo No. 2 Hospital , No. 41 Northwest Street , Ningbo , Zhejiang Province 315010 , China
| | - Xinwei Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Qin Du
- Department of Gastroenterology, The Second Affiliated Hospital, College of Medicine , Zhejiang University , Hangzhou , Zhejiang Province 310009 , China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy , Fudan University , Shanghai 201203 , China
| |
Collapse
|
32
|
Abstract
Pelvic ultrasound examination is the primary imaging modality for evaluating a wide range of female pelvic symptomatology, and is often the first imaging test to detect a gynecologic malignancy. Ultrasound imaging is particularly useful for evaluating the thickness and appearance of the endometrium in patients with abnormal bleeding, and in detecting and characterizing ovarian lesions. This article reviews the ultrasound appearance of gynecologic neoplasms grouped by anatomic site of origin, the ultrasound appearance of select benign pelvic pathology not to be misinterpreted as malignancy, as well as available ultrasound imaging-based guidelines for managing potential gynecologic neoplasms.
Collapse
|
33
|
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
|
34
|
Qiu C, Yin T, Zhang Y, Lian Y, You Y, Wang K, Zheng R, Shuai X. Ultrasound Imaging Based on Molecular Targeting for Quantitative Evaluation of Hepatic Ischemia-Reperfusion Injury. Am J Transplant 2017; 17:3087-3097. [PMID: 28489274 DOI: 10.1111/ajt.14345] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 01/25/2023]
Abstract
The aim of the present study was to quantitatively diagnose and monitor the therapy response of hepatic ischemia-reperfusion injury (IRI) with the use of targeted ultrasound (US) imaging. Targeted microbubbles (MBs) were fabricated, and the binding of intracellular adhesion molecule 1 (ICAM-1) antibodies to MBs was observed. To establish a quantitative method based on targeted US imaging, contrast-enhanced US was applied for IRI rats. After andrographolide treatment, the IRI rats were subjected to the quantitative targeted US imaging for a therapeutic effect. Effective binding of ICAM-1 antibodies to MBs was observed. According to the quantitative targeted US imaging, the ICAM-1 normalized intensity difference (NID) in the IRI rats (38.74 ± 15.08%) was significantly higher than that in the control rats (10.08 ± 2.52%, p = 0.048). Further, different degrees of IRI (mild IRI, moderate to severe IRI) were distinguished by the use of the NID (37.14 ± 2.14%, 22.34 ± 1.08%, p = 0.002). Analysis of mRNA expression demonstrated the accuracy of analyzing the NID by using quantitative targeted US imaging (R2 = 0.7434, p < 0.001). Andrographolide treatment resulted in an obviously weakened NID of ICAM-1 (17.7 ± 4.8% vs 34.2 ± 6.6%, p < 0.001). The study showed the potential of the quantitative targeted US imaging method for the diagnosis and therapeutic monitoring of IRI.
Collapse
Affiliation(s)
- C Qiu
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - T Yin
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Y Zhang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Y Lian
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Y You
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - K Wang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Science, Beijing, China
| | - R Zheng
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - X Shuai
- PCFM Lab of Ministry of Education, School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou, China.,Center for Biomedical Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
The Use of Acoustic Radiation Force Decorrelation-Weighted Pulse Inversion for Enhanced Ultrasound Contrast Imaging. Invest Radiol 2017; 52:95-102. [PMID: 27495188 DOI: 10.1097/rli.0000000000000313] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES The use of ultrasound imaging for cancer diagnosis and screening can be enhanced with the use of molecularly targeted microbubbles. Nonlinear imaging strategies such as pulse inversion (PI) and "contrast pulse sequences" (CPS) can be used to differentiate microbubble signal, but often fail to suppress highly echogenic tissue interfaces. This failure results in false-positive detection and potential misdiagnosis. In this study, a novel acoustic radiation force (ARF)-based approach was developed for superior microbubble signal detection. The feasibility of this technique, termed ARF decorrelation-weighted PI (ADW-PI), was demonstrated in vivo using a subcutaneous mouse tumor model. MATERIALS AND METHODS Tumors were implanted in the hindlimb of C57BL/6 mice by subcutaneous injection of MC38 cells. Lipid-shelled microbubbles were conjugated to anti-VEGFR2 antibody and administered via bolus injection. An image sequence using ARF pulses to generate microbubble motion was combined with PI imaging on a Verasonics Vantage programmable scanner. ADW-PI images were generated by combining PI images with interframe signal decorrelation data. For comparison, CPS images of the same mouse tumor were acquired using a Siemens Sequoia clinical scanner. RESULTS Microbubble-bound regions in the tumor interior exhibited significantly higher signal decorrelation than static tissue (n = 9, P < 0.001). The application of ARF significantly increased microbubble signal decorrelation (n = 9, P < 0.01). Using these decorrelation measurements, ADW-PI imaging demonstrated significantly improved microbubble contrast-to-tissue ratio when compared with corresponding CPS or PI images (n = 9, P < 0.001). Contrast-to-tissue ratio improved with ADW-PI by approximately 3 dB compared with PI images and 2 dB compared with CPS images. CONCLUSIONS Acoustic radiation force can be used to generate adherent microbubble signal decorrelation without microbubble bursting. When combined with PI, measurements of the resulting microbubble signal decorrelation can be used to reconstruct images that exhibit superior suppression of highly echogenic tissue interfaces when compared with PI or CPS alone.
Collapse
|
36
|
Volz KR, Evans KD, Kanner CD, Buford JA, Freimer M, Sommerich CM, Basso DM. Molecular Ultrasound Imaging for the Detection of Neural Inflammation: A Longitudinal Dosing Pilot Study. JOURNAL OF DIAGNOSTIC MEDICAL SONOGRAPHY 2017. [DOI: 10.1177/8756479317736250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Molecular ultrasound imaging provides the ability to detect physiologic processes noninvasively by targeting a variety of biomarkers in vivo. The current study was performed by exploiting an inflammatory biomarker, P-selectin, known to be present following spinal cord injury. Using a murine model (n = 6), molecular ultrasound imaging was performed using contrast microbubbles modified to target and adhere to P-selectin, prior to spinal cord injury (0D), acute stage postinjury (7D), and chronic stage (42D). Additionally, two imaging sessions were performed on each subject at specific time points, using doses of 30 μL and 100 μL. Upon analysis, targeted contrast analysis parameters were appreciably increased during the 7D scan compared with the 42D scan, without statistical significance. When examining the dose levels, the 30-μL dose demonstrated greater values than the 100-μL dose but lacked statistical significance. These findings provide additional preclinical evidence for the use of molecular ultrasound imaging for the possible detection of inflammation.
Collapse
Affiliation(s)
- Kevin R. Volz
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Kevin D. Evans
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - John A. Buford
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Miriam Freimer
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - D. Michele Basso
- College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
37
|
Nilotinib Enhances Tumor Angiogenesis and Counteracts VEGFR2 Blockade in an Orthotopic Breast Cancer Xenograft Model with Desmoplastic Response. Neoplasia 2017; 19:896-907. [PMID: 28938160 PMCID: PMC5608503 DOI: 10.1016/j.neo.2017.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/14/2022] Open
Abstract
Vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR)-targeted therapies predominantly affect nascent, immature tumor vessels. Since platelet-derived growth factor receptor (PDGFR) blockade inhibits vessel maturation and thus increases the amount of immature tumor vessels, we evaluated whether the combined PDGFR inhibition by nilotinib and VEGFR2 blockade by DC101 has synergistic therapy effects in a desmoplastic breast cancer xenograft model. In this context, besides immunohistological evaluation, molecular ultrasound imaging with BR55, the clinically used VEGFR2-targeted microbubbles, was applied to monitor VEGFR2-positive vessels noninvasively and to assess the therapy effects on tumor angiogenesis. DC101 treatment alone inhibited tumor angiogenesis, resulting in lower tumor growth and in significantly lower vessel density than in the control group after 14 days of therapy. In contrast, nilotinib inhibited vessel maturation but enhanced VEGFR2 expression, leading to markedly increased tumor volumes and a significantly higher vessel density. The combination of both drugs led to an almost similar tumor growth as in the DC101 treatment group, but VEGFR2 expression and microvessel density were higher and comparable to the controls. Further analyses revealed significantly higher levels of tumor cell–derived VEGF in nilotinib-treated tumors. In line with this, nilotinib, especially in low doses, induced an upregulation of VEGF and IL-6 mRNA in the tumor cells in vitro, thus providing an explanation for the enhanced angiogenesis observed in nilotinib-treated tumors in vivo. These findings suggest that nilotinib inhibits vessel maturation but counteracts the effects of antiangiogenic co-therapy by enhancing VEGF expression by the tumor cells and stimulating tumor angiogenesis.
Collapse
|
38
|
Pang T, Liu ZD, Deng K, Zhang CQ, Wang GL. Preliminary application of multiple parameters spectral CT in the diagnosis of ovarian cancer. Medicine (Baltimore) 2017; 96:e7786. [PMID: 28796078 PMCID: PMC5556244 DOI: 10.1097/md.0000000000007786] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The aim of the study was to evaluate the effectiveness of spectral CT in the diagnosis of ovarian cancer. We retrospectively analyzed the data of 22 patients with spectral CT enhanced scan. The patients were divided into 2 groups: ovarian cancer group (n = 11) and benign tumor group (n = 11), according to the pathologic results. CT values at 40 keV, iodine concentration (IC), water concentration (WC) and spectral curve slope (λHU) of arterial phase and venous phase in the tumors of 2 groups were measured with gemstone spectral imaging (GSI) post-processing software. The independent samples t test was used to compare these multiple parameters above between 2 groups. For the parameters which showed statistically different, the ROC curves were further generated to calculate their diagnostic effectiveness respectively. According to the results, CT values at 40 keV, IC and λHU measured in arterial and venous phases were higher in ovarian cancer group than those in benign tumor group. There were significant differences between these 2 groups (P < 0.05). While WC had no significant difference in these 2 groups (P > 0.05). CT values at 40 keV, IC and λHU had high effectiveness to the diagnosis of ovarian cancer according to ROC curves. The optimal parameter among them was IC in arterial phase with AUC of 0.90. Using 10.92 (100 ug/cm3) as a threshold value, the sensitivity and specificity for diagnosis were 88.9% and 94.7%. Thus, we concluded that spectral CT with multiple parameters was valuable in the diagnosis of ovarian cancer.
Collapse
|
39
|
Wischhusen J, Padilla F. Microbubble Enzyme-Linked Immunosorbent Assay for the Detection of Targeted Microbubbles in in Vitro Static Binding Assays. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:1506-1519. [PMID: 28450034 DOI: 10.1016/j.ultrasmedbio.2017.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/28/2017] [Accepted: 03/07/2017] [Indexed: 06/07/2023]
Abstract
Targeted microbubbles (MBs) are ultrasound contrast agents that are functionalized with a ligand for ultrasound molecular imaging of endothelial markers. Novel targeted MBs are characterized in vitro by incubation in protein-coated wells, followed by binding quantification by microscopy or ultrasound imaging. Both methods provide operator-dependent results: Between 3 and 20 fields of view from a heterogeneous sample are typically selected for analysis by microscopy, and in ultrasound imaging, different acoustic settings affect signal intensities. This study proposes a new method to reproducibly quantify MB binding based on enzyme-linked immunosorbent assay (ELISA), in which bound MBs are revealed with an enzyme-linked antibody. MB-ELISA was adapted to in vitro static binding assays, incubating the MBs in inverted position or by agitation, and compared with microscopy. The specificity and sensitivity of MB-ELISA enable the reliable quantification of MB binding in a rapid, high-throughput and whole-well analysis, facilitating the characterization of new targeted contrast agents.
Collapse
Affiliation(s)
| | - Frederic Padilla
- INSERM, U1032, LabTAU, Lyon, France; Université de Lyon, Lyon, France.
| |
Collapse
|
40
|
Mullick Chowdhury S, Lee T, Willmann JK. Ultrasound-guided drug delivery in cancer. Ultrasonography 2017; 36:171-184. [PMID: 28607323 PMCID: PMC5494871 DOI: 10.14366/usg.17021] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/23/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022] Open
Abstract
Recent advancements in ultrasound and microbubble (USMB) mediated drug delivery technology has shown that this approach can improve spatially confined delivery of drugs and genes to target tissues while reducing systemic dose and toxicity. The mechanism behind enhanced delivery of therapeutics is sonoporation, the formation of openings in the vasculature, induced by ultrasound-triggered oscillations and destruction of microbubbles. In this review, progress and challenges of USMB mediated drug delivery are summarized, with special focus on cancer therapy.
Collapse
Affiliation(s)
| | - Taehwa Lee
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jürgen K. Willmann
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
41
|
Ma YY, Jin KT, Wang SB, Wang HJ, Tong XM, Huang DS, Mou XZ. Molecular Imaging of Cancer with Nanoparticle-Based Theranostic Probes. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:1026270. [PMID: 29097909 PMCID: PMC5612740 DOI: 10.1155/2017/1026270] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/16/2017] [Indexed: 11/18/2022]
Abstract
Although advancements in medical technology supporting cancer diagnosis and treatment have improved survival, these technologies still have limitations. Recently, the application of noninvasive imaging for cancer diagnosis and therapy has become an indispensable component in clinical practice. However, current imaging contrasts and tracers, which are in widespread clinical use, have their intrinsic limitations and disadvantages. Nanotechnologies, which have improved in vivo detection and enhanced targeting efficiency for cancer, may overcome some of the limitations of cancer diagnosis and therapy. Theranostic nanoparticles have great potential as a therapeutic model, which possesses the ability of their nanoplatforms to load targeted molecule for both imaging and therapeutic functions. The resulting nanosystem will likely be critical with the growth of personalized medicine because of their diagnostic potential, effectiveness as a drug delivery vehicle, and ability to oversee patient response to therapy. In this review, we discuss the achievements of modern nanoparticles with the goal of accurate tumor imaging and effective treatment and discuss the future prospects.
Collapse
Affiliation(s)
- Ying-Yu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
- Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province, Hangzhou 310014, China
| | - Ke-Tao Jin
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, China
| | - Shi-Bing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
- Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province, Hangzhou 310014, China
| | - Hui-Ju Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
- Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province, Hangzhou 310014, China
| | - Xiang-Min Tong
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
- Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province, Hangzhou 310014, China
| | - Dong-Sheng Huang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
- Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province, Hangzhou 310014, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
- Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province, Hangzhou 310014, China
- School of Basic Medical Sciences, Hangzhou Medical College, Hangzhou 310053, China
| |
Collapse
|
42
|
Contrast-enhanced ultrasound evaluation of pancreatic cancer xenografts in nude mice after irradiation with sub-threshold focused ultrasound for tumor ablation. Oncotarget 2017; 8:37584-37593. [PMID: 28402267 PMCID: PMC5514932 DOI: 10.18632/oncotarget.16621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/09/2017] [Indexed: 01/15/2023] Open
Abstract
We evaluated the efficacy of contrast-enhanced ultrasound for assessing tumors after irradiation with sub-threshold focused ultrasound (FUS) ablation in pancreatic cancer xenografts in nude mice. Thirty tumor-bearing nude mice were divided into three groups: Group A received sham irradiation, Group B received a moderate-acoustic energy dose (sub-threshold), and Group C received a high-acoustic energy dose. In Group B, B-mode ultrasound (US), color Doppler US, and dynamic contrast-enhanced ultrasound (DCE-US) studies were conducted before and after irradiation. After irradiation, tumor growth was inhibited in Group B, and the tumors shrank in Group C. In Group A, the tumor sizes were unchanged. In Group B, contrast-enhanced ultrasound (CEUS) images showed a rapid rush of contrast agent into and out of tumors before irradiation. After irradiation, CEUS revealed contrast agent perfusion only at the tumor periphery and irregular, un-perfused volumes of contrast agent within the tumors. DCE-US perfusion parameters, including peak intensity (PI) and area under the curve (AUC), had decreased 24 hours after irradiation. PI and AUC were increased 48 hours and 2weeks after irradiation. Time to peak (TP) and sharpness were increased 24 hours after irradiation. TP decreased at 48 hours and 2 weeks after irradiation. CEUS is thus an effective method for early evaluation after irradiation with sub-threshold FUS.
Collapse
|
43
|
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]
|
44
|
Lee HJ, Hwang SI, Byun J, Kong HY, Jung HS, Kang M. Ultrasound contrast-enhanced study as an imaging biomarker for anti-cancer drug treatment: preliminary study with paclitaxel in a xenograft mouse tumor model (secondary publication). Ultrasonography 2017; 36:370-377. [PMID: 28290184 PMCID: PMC5621803 DOI: 10.14366/usg.17015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/10/2017] [Accepted: 02/14/2017] [Indexed: 11/03/2022] Open
Abstract
PURPOSE The purpose of this study was to assess tumor angiogenesis using contrast-enhanced ultrasonography (CEUS) of human prostate cancer cells (PC3) that were implanted in mice before and after paclitaxel injection. METHODS Twelve mice were injected with human PC3. The mice were grouped into two groups; one was the paclitaxel-treated group (n=6) and the other was the control group (n=6). Before administering paclitaxel into the peritoneal cavity, baseline CEUS was performed after the administration of 500 μL (1×108 microbubbles) of contrast agent. The area under the curve (AUC) up to 50 seconds after injection was derived from the time-intensity curves. After injection of paclitaxel or saline, CEUS studies were performed at the 1-week follow-up. Changes in tumor volume and the AUC in both two groups were evaluated. After CEUS, the microvessel density (MVD) was compared between the groups. RESULTS In the paclitaxel-treated group, the AUC from CEUS showed a significant decrease 1-week after paclitaxel administration (P=0.030), even though the tumor volume showed no significant changes (P=0.116). In the control group, there was no significant decrease of the AUC (P=0.173). Pathologically, there was a significant difference in MVD between both groups (P=0.002). CONCLUSION The AUC from the time intensity curve derived from CEUS showed an early change in response to the anti-cancer drug treatment that preceded the change in tumor size. The findings of CEUS could serve as an imaging biomarker for assessing tumor responses to anti-cancer drug treatment.
Collapse
Affiliation(s)
- Hak Jong Lee
- Department of Radiology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sung Il Hwang
- Department of Radiology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jonghoe Byun
- Department of Molecular Biology, Dankook University, Cheonan, Korea
| | - Hoon Young Kong
- Department of Molecular Biology, Dankook University, Cheonan, Korea
| | - Hyun Sook Jung
- Department of Radiology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, Korea
| | - Mira Kang
- Department of Radiology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
45
|
Lee HJ, Yoon TJ, Yoon YI. Synthesis of ultrasound contrast agents: characteristics and size distribution analysis (secondary publication). Ultrasonography 2017; 36:378-384. [PMID: 28290183 PMCID: PMC5621802 DOI: 10.14366/usg.17014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/10/2017] [Accepted: 02/14/2017] [Indexed: 11/03/2022] Open
Abstract
PURPOSE The purpose of this study was to establish a method for ultrasound (US) contrast agent synthesis and to evaluate the characteristics of the synthesized US contrast agent. METHODS A US contrast agent, composed of liposome and sulfur hexafluoride (SF6), was synthesized by dissolving 21 μmol 1,2-dihexadecanoyl-sn-glycero-3-phosphocholine (DPPC, C40H80NO8P), 9 μmol cholesterol, and 1.9 μmol of dihexadecylphosphate (DCP, [CH3(CH2)15O]2P(O)OH) in chloroform. After evaporation in a warm water bath and drying for 12-24 hours, the contrast agent was synthesized using the sonication process by the addition of a buffer and SF6 gas. The size distribution of the bubbles was analyzed using dynamic light scattering measurement methods. The degradation curve was evaluated by assessing the change in the number of contrast agent bubbles using light microscopy immediately, 12, 24, 36, 48, 60, 72, and 84 hours after synthesis. The echogenicity of the synthesized microbubbles was compared with commercially available microbubbles (SonoVue, Bracco). RESULTS contrast agent was synthesized successfully using an evaporation-drying-sonication method. Most bubbles had a mean diameter of 154.2 nm and showed marked degradation 24 hours after synthesis. Although no statistically significant differences were observed between SonoVue and the synthesized contrast agent, a difference in echogenicity was observed between the synthesized contrast agent and saline (P<0.01). CONCLUSION We successfully synthesized a US contrast agent using an evaporation-dryingsonication method. These results may help future research in the fields of anticancer drug delivery, gene delivery, targeted molecular imaging, and targeted therapy.
Collapse
Affiliation(s)
- Hak Jong Lee
- Program in Nano Science and Technology, Department of Transdisciplinary Studies, Seoul National University Graduate School of Convergence Science and Technology, Seoul, Korea.,Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Tae-Jong Yoon
- Department of Applied Bioscience, CHA University, Pocheon, Korea
| | - Young Il Yoon
- Program in Nano Science and Technology, Department of Transdisciplinary Studies, Seoul National University Graduate School of Convergence Science and Technology, Seoul, Korea
| |
Collapse
|
46
|
Zhang H, Ingham ES, Gagnon MKJ, Mahakian LM, Liu J, Foiret JL, Willmann JK, Ferrara KW. In vitro characterization and in vivo ultrasound molecular imaging of nucleolin-targeted microbubbles. Biomaterials 2017; 118:63-73. [PMID: 27940383 PMCID: PMC5279957 DOI: 10.1016/j.biomaterials.2016.11.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/11/2016] [Accepted: 11/20/2016] [Indexed: 12/12/2022]
Abstract
Nucleolin (NCL) plays an important role in tumor vascular development. An increased endothelial expression level of NCL has been related to cancer aggressiveness and prognosis and has been detected clinically in advanced tumors. Here, with a peptide targeted to NCL (F3 peptide), we created an NCL-targeted microbubble (MB) and compared the performance of F3-conjugated MBs with non-targeted (NT) MBs both in vitro and in vivo. In an in vitro study, F3-conjugated MBs bound 433 times more than NT MBs to an NCL-expressing cell line, while pretreating cells with 0.5 mM free F3 peptide reduced the binding of F3-conjugated MBs by 84%, n = 4, p < 0.001. We then set out to create a method to extract both the tumor wash-in and wash-out kinetics and tumor accumulation following a single injection of targeted MBs. In order to accomplish this, a series of ultrasound frames (a clip) was recorded at the time of injection and subsequent time points. Each pixel within this clip was analyzed for the minimum intensity projection (MinIP) and average intensity projection (AvgIP). We found that the MinIP robustly demonstrates enhanced accumulation of F3-conjugated MBs over the range of tumor diameters evaluated here (2-8 mm), and the difference between the AvgIP and the MinIP quantifies inflow and kinetics. The inflow and clearance were similar for unbound F3-conjugated MBs, control (non-targeted) and scrambled control agents. Targeted agent accumulation was confirmed by a high amplitude pulse and by a two-dimensional Fourier Transform technique. In summary, F3-conjugated MBs provide a new imaging agent for ultrasound molecular imaging of cancer vasculature, and we have validated metrics to assess performance using low mechanical index strategies that have potential for use in human molecular imaging studies.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | - M Karen J Gagnon
- Department of Environmental Health and Safety, University of California, Davis, CA, 95616, USA
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | - Jingfei Liu
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | - Josquin L Foiret
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | | | - Katherine W Ferrara
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
47
|
Abstract
Pancreatic diseases, chronic pancreatitis, pancreatic cancer and diabetes mellitus, taken together, occur in >10% of the world population. Pancreatic diseases, as with other diseases, benefit from early intervention and appropriate diagnosis. Although imaging technologies have given clinicians an unprecedented toolbox to aid in clinical decision-making, advances in these technologies and development of molecular-based diagnostic tools could enable physicians to identify diseases at an even earlier stage and, thereby, improve patient outcomes. In this Review, we discuss and identify gaps in the use of imaging techniques for the early detection and appropriate treatment stratification of various pancreatic diseases, including diabetes mellitus, acute and chronic pancreatitis and pancreatic cancer. Imaging techniques discussed are MRI, CT, PET and ultrasonography. Additionally, the identification of new molecular targets for imaging and the development of contrast agents that are able to give molecular information in noninvasive radionuclear imaging and ultrasonography are emerging areas of innovation that could lead to increased diagnostic accuracy and improved patient outcomes.
Collapse
Affiliation(s)
- Julien Dimastromatteo
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Building MR5, Charlottesville, Virginia 22903, USA
| | - Teresa Brentnall
- Division of Gastroenterology, Digestive Diseases Center, 1959 Northeast Pacific Street, Seattle, Washington 98195, USA
| | - Kimberly A Kelly
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Building MR5, Charlottesville, Virginia 22903, USA
| |
Collapse
|
48
|
Volz KR, Evans KD, Kanner CD, Buford JA, Freimer M, Sommerich CM. Targeted Contrast-Enhanced Ultrasound for Inflammation Detection. JOURNAL OF DIAGNOSTIC MEDICAL SONOGRAPHY 2016. [DOI: 10.1177/8756479316678616] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Molecular imaging is a form of nanotechnology that enables the noninvasive examination of biological processes in vivo. Radiopharmaceutical agents are used to target biochemical markers, permitting their detection and evaluation. Early visualization of molecular variations indicative of pathophysiological processes can aid in patient diagnoses and management decisions. Molecular imaging is performed by introducing into the body molecular probes, which are often contrast agents that have been nanoengineered to target and tether to molecules, thus enabling their radiologic identification. Through a nanoengineering process, ultrasound contrast agents can be targeted to specific molecules, extending ultrasound’s capabilities from the tissue to molecular level. Molecular ultrasound, or targeted contrast-enhanced ultrasound (TCEUS), has recently emerged as a popular molecular imaging technique due to its ability to provide real-time anatomic and functional information without ionizing radiation. However, molecular ultrasound represents a novel form of molecular imaging and consequently remains largely preclinical. This review explores the commonalities of TCEUS across several molecular targets and points to the need for standardization of kinetic behavior analysis. The literature underscores evidence gaps and the need for additional research. The application of TCEUS is unlimited but needs further standardization to ensure that future research studies are comparable.
Collapse
Affiliation(s)
- Kevin R. Volz
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| | - Kevin D. Evans
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| | - Christopher D. Kanner
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| | - John A. Buford
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| | - Miriam Freimer
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| | | |
Collapse
|
49
|
de Geus SWL, Boogerd LSF, Swijnenburg RJ, Mieog JSD, Tummers WSFJ, Prevoo HAJM, Sier CFM, Morreau H, Bonsing BA, van de Velde CJH, Vahrmeijer AL, Kuppen PJK. Selecting Tumor-Specific Molecular Targets in Pancreatic Adenocarcinoma: Paving the Way for Image-Guided Pancreatic Surgery. Mol Imaging Biol 2016; 18:807-819. [PMID: 27130234 PMCID: PMC5093212 DOI: 10.1007/s11307-016-0959-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE The purpose of this study was to identify suitable molecular targets for tumor-specific imaging of pancreatic adenocarcinoma. PROCEDURES The expression of eight potential imaging targets was assessed by the target selection criteria (TASC)-score and immunohistochemical analysis in normal pancreatic tissue (n = 9), pancreatic (n = 137), and periampullary (n = 28) adenocarcinoma. RESULTS Integrin αvβ6, carcinoembryonic antigen (CEA), epithelial growth factor receptor (EGFR), and urokinase plasminogen activator receptor (uPAR) showed a significantly higher (all p < 0.001) expression in pancreatic adenocarcinoma compared to normal pancreatic tissue and were confirmed by the TASC score as promising imaging targets. Furthermore, these biomarkers were expressed in respectively 88 %, 71 %, 69 %, and 67 % of the pancreatic adenocarcinoma patients. CONCLUSIONS The results of this study show that integrin αvβ6, CEA, EGFR, and uPAR are suitable targets for tumor-specific imaging of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Susanna W L de Geus
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Leonora S F Boogerd
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Rutger-Jan Swijnenburg
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Willemieke S F J Tummers
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Hendrica A J M Prevoo
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Cornelis J H van de Velde
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
50
|
Cui Y, Zhang C, Luo R, Liu H, Zhang Z, Xu T, Zhang Y, Wang D. Noninvasive monitoring of early antiangiogenic therapy response in human nasopharyngeal carcinoma xenograft model using MRI with RGD-conjugated ultrasmall superparamagnetic iron oxide nanoparticles. Int J Nanomedicine 2016; 11:5671-5682. [PMID: 27895477 PMCID: PMC5117895 DOI: 10.2147/ijn.s115357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose Arginine-glycine-aspartic acid (RGD)-based nanoprobes allow specific imaging of integrin αvβ3, a protein overexpressed during angiogenesis. Therefore, this study applied a novel RGD-coupled, polyacrylic acid (PAA)-coated ultrasmall superparamagnetic iron oxide (USPIO) (referred to as RGD-PAA-USPIO) in order to detect tumor angiogenesis and assess the early response to antiangiogenic treatment in human nasopharyngeal carcinoma (NPC) xenograft model by magnetic resonance imaging (MRI). Materials and methods The binding specificity of RGD-PAA-USPIO with human umbilical vein endothelial cells (HUVECs) was confirmed by Prussian blue staining and transmission electron microscopy in vitro. The tumor targeting of RGD-PAA-USPIO was evaluated in the NPC xenograft model. Later, mice bearing NPC underwent MRI at baseline and after 4 and 14 days of consecutive treatment with Endostar or phosphate-buffered saline (n=10 per group). Results The specific uptake of the RGD-PAA-USPIO nanoparticles was mainly dependent on the interaction between RGD and integrin αvβ3 of HUVECs. The tumor targeting of RGD-PAA-USPIO was observed in the NPC xenograft model. Moreover, the T2 relaxation time of mice in the Endostar-treated group decreased significantly compared with those in the control group both on days 4 and 14, consistent with the immunofluorescence results of CD31 and CD61 (P<0.05). Conclusion This study demonstrated that the magnetic resonance molecular nanoprobes, RGD-PAA-USPIOs, allow noninvasive in vivo imaging of tumor angiogenesis and assessment of the early response to antiangiogenic treatment in NPC xenograft model, favoring its potential clinical translation.
Collapse
Affiliation(s)
- Yanfen Cui
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Caiyuan Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Ran Luo
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Huanhuan Liu
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Zhongyang Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Tianyong Xu
- MR Advanced Application and Research Center, GE Healthcare China, Shanghai, People's Republic of China
| | - Yong Zhang
- MR Advanced Application and Research Center, GE Healthcare China, Shanghai, People's Republic of China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| |
Collapse
|