1
|
Gerena M, Allen BC, Turkbey B, Barker SJ, Costa DN, Flink C, Meyers ML, Ramasamy R, Rosario J, Sharma A, Whitworth P, Williams WL, Oto A. ACR Appropriateness Criteria® Acute Onset of Scrotal Pain-Without Trauma, Without Antecedent Mass: 2024 Update. J Am Coll Radiol 2024; 21:S364-S371. [PMID: 39488348 DOI: 10.1016/j.jacr.2024.08.011] [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: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 11/04/2024]
Abstract
Acute scrotum is a medical emergency that requires prompt accurate diagnosis to appropriately triage potentially surgical conditions. Numerous differential diagnoses with overlapping clinical presentations make this a diagnostic challenge. Ultrasound is the established first-line imaging modality for acute scrotal disease and can be used to diagnose most scrotal disorders promptly and with high accuracy. The American College of Radiology Appropriateness Criteria are evidence-based guidelines for specific clinical conditions that are reviewed annually by a multidisciplinary expert panel. The guideline development and revision process support the systematic analysis of the medical literature from peer reviewed journals. Established methodology principles such as Grading of Recommendations Assessment, Development, and Evaluation or GRADE are adapted to evaluate the evidence. The RAND/UCLA Appropriateness Method User Manual provides the methodology to determine the appropriateness of imaging and treatment procedures for specific clinical scenarios. In those instances where peer reviewed literature is lacking or equivocal, experts may be the primary evidentiary source available to formulate a recommendation.
Collapse
Affiliation(s)
- Marielia Gerena
- Stony Brook University, Renaissance School of Medicine, Stony Brook, New York.
| | - Brian C Allen
- Panel Chair, Duke University Medical Center, Durham, North Carolina
| | - Baris Turkbey
- Panel Vice-Chair, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | - Carl Flink
- University of Cincinnati, Cincinnati, Ohio; Committee on Emergency Radiology-GSER
| | - Mariana L Meyers
- Children's Hospital Colorado. University of Colorado School of Medicine, Aurora, Colorado
| | - Ranjith Ramasamy
- University of Miami Miller School of Medicine, Miami, Florida; American Urological Association
| | - Javier Rosario
- HCA Florida Osceola Hospital, Kissimmee, Florida; American College of Emergency Physicians
| | - Akash Sharma
- Mayo Clinic, Jacksonville, Florida; Commission on Nuclear Medicine and Molecular Imaging
| | - Pat Whitworth
- Thomas F. Frist, Jr College of Medicine, Belmont University, Nashville, Tennessee
| | - Winter L Williams
- AB Heersink School of Medicine, Birmingham, Alabama, Internal medicine
| | - Aytekin Oto
- Specialty Chair, University of Chicago, Chicago, Illinois
| |
Collapse
|
2
|
McCorkell G, Piva T, Highgate D, Nakayama M, Geso M. Ultrasound-stimulated microbubbles to enhance radiotherapy: A scoping review. J Med Imaging Radiat Oncol 2024; 68:740-769. [PMID: 39250692 DOI: 10.1111/1754-9485.13740] [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: 02/24/2024] [Accepted: 07/22/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Primarily used as ultrasound contrast agents, microbubbles have recently emerged as a versatile therapeutic vector that can be 'burst' to deliver payloads in the presence of suitably optimised ultrasound fields. Ultrasound-stimulated microbubbles (USMB) have recently demonstrated improvements in treatment outcomes across a variety of clinical applications. This scoping review investigates whether this potential translates into the context of radiation therapy by evaluating the application of this technology across all three phases of radiation action. METHODS Primary research articles, excluding poster presentations and conference proceedings, were identified through systematic searches of the PubMed NCBI/Medline, Embase/OVID, Web of Science and CINAHL/EBSCOhost databases, with additional articles identified via manual Google Scholar searching. Articles were dual screened for inclusion using the Covidence systematic review platform and classified against all three phases of radiation action. RESULTS Overall, 57 eligible publications from a total of 1389 identified articles were included in the review, with studies dating back to 2012. Study heterogeneity prevented formal statistical analysis; however, most articles reported improved outcomes using USMB in the presence of radiation compared to that of radiation alone. These improvements appear to result from the use of USMB as either a biovascular disruptor causing tumour cell damage via indirect mechanisms, or as a localised treatment vector that directly increases tumour cell uptake of other therapeutic and physical agents designed to enhance radiation action. CONCLUSIONS USMB demonstrate exciting potential to enhance the effects of radiation treatments due to their versatility and capacity to target all three phases of radiation action.
Collapse
Affiliation(s)
- Giulia McCorkell
- RMIT University, Melbourne, Victoria, Australia
- The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | - Masao Nakayama
- RMIT University, Melbourne, Victoria, Australia
- Kobe University, Kobe, Hyogo, Japan
- Kita-Harima Medical Center, Ono, Hyogo, Japan
| | - Moshi Geso
- RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Oglat AA. A review of ultrasound contrast media. F1000Res 2024; 12:1444. [PMID: 38817410 PMCID: PMC11137482 DOI: 10.12688/f1000research.140131.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/03/2024] [Indexed: 06/01/2024] Open
Abstract
Efforts have been made over the last five decades to create effective ultrasonic contrast media (UCM) for cardiac and noncardiac applications. The initial UCM was established in the 1980s, following publications from the 1960s that detailed the discovery of ultrasonic contrast enhancement using small gaseous bubbles in echocardiographic examinations. An optimal contrast agent for echography should possess the following characteristics: non-toxicity, suitability for intravenous injection, ability to traverse pulmonary, cardiac, and capillary circulations, and stability for recirculation. Definity, Optison, Sonazoid, and SonoVue are examples of current commercial contrast media. These contrast media have shown potential for various clinical reasons, both on-label and off-label. Several possible UCMs have been developed or are in progress. Advancements in comprehending the physical, chemical, and biological characteristics of microbubbles have significantly improved the visualization of tumor blood vessels, the identification of areas with reduced blood supply, and the enhanced detection of narrowed blood vessels. Innovative advances are expected to enhance future applications such as ultrasonic molecular imaging and therapeutic utilization of microbubbles.
Collapse
Affiliation(s)
- Ammar A. Oglat
- Department of Medical Imaging, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan., The Hashemite University, Az-Zarqa, Zarqa Governorate, 13133, Jordan
| |
Collapse
|
4
|
Hosseini RS, Ebrahimi PS, Shokrani A. Diagnostic value of enhanced-contrast ultrasound for cesarean scar pregnancy: A systematic review. Eur J Obstet Gynecol Reprod Biol 2024; 293:119-124. [PMID: 38145593 DOI: 10.1016/j.ejogrb.2023.12.028] [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: 09/27/2023] [Revised: 12/13/2023] [Accepted: 12/17/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND Cesarean scar pregnancy, an uncommon ectopic pregnancy in which the embryo is implanted in the cesarean scar, poses significant risks without prompt diagnosis and treatment. Its prevalence has risen alongside increased cesarean section rates. Despite various treatment approaches, consensus remains elusive. Ultrasonography, particularly contrast-enhanced ultrasonography, shows promise in cesarean scar pregnancy diagnosis. MAIN BODY This systematic review, following PRISMA guidelines, explores contrast-enhanced ultrasound's diagnostic potential in cesarean scar pregnancy. We searched PubMed, Scopus, Web of Science, and Google Scholar up to August 2023. Selection involved two stages: title/abstract screening and full-text assessment. The included studies investigated contrast-enhanced ultrasound's diagnostic value in cesarean scar pregnancy, provided adequate data, and were peer-reviewed in English. Quality assessment followed the QUADAS-2 criteria. We extracted the diagnostic accuracy metrics: sensitivity, specificity, and accuracy. Out of 193 records, five studies met the inclusion criteria (2016-2020, China). Contrast-enhanced ultrasound displayed sensitivities of 77%-100% and specificities of 95%-100%. Two studies reported accuracy of 96.9%-97.8%. Compared with conventional ultrasound, contrast-enhanced ultrasound exhibited superior sensitivity, specificity, and accuracy. It also outperformed transvaginal ultrasound. CONCLUSION Enhanced-contrast ultrasound holds promise for diagnosing and managing cesarean scar pregnancy by visualizing scar vascularization in real-time, thereby reducing severe complication risks. This review highlights contrast-enhanced ultrasound as a transformative diagnostic tool for cesarean scar pregnancy management, despite existing evidence limitations.
Collapse
Affiliation(s)
- Reza Shah Hosseini
- Istanbul Medipol University, Faculty of Medicine, Medical Student, Istanbul, Turkey.
| | | | - Aniseh Shokrani
- Istanbul Medipol University, Faculty of Medicine, Medical Student, Istanbul, Turkey
| |
Collapse
|
5
|
Polydorou AE, May JP, Makris K, Ferri S, Wu Q, Stride E, Carugo D, Evans ND. An investigation into the cytotoxic effects of microbubbles and their constituents on osteosarcoma and bone marrow stromal cells. Biochim Biophys Acta Gen Subj 2023; 1867:130481. [PMID: 37802372 DOI: 10.1016/j.bbagen.2023.130481] [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: 07/19/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Ultrasound-responsive microbubbles offer a means of achieving minimally invasive, localised drug delivery in applications including regenerative medicine. To facilitate their use, however, it is important to determine any cytotoxic effects they or their constituents may have. The aim of this study was to test the hypothesis that phospholipid-shelled microbubbles are non-toxic to human bone-derived cells at biologically-relevant concentrations. METHODS Microbubbles were fabricated using combinations of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dibehenoyl-sn-glycero-3-phosphocholine (DBPC), polyoxyethylene(40) stearate (PEG40S) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene-glycol)-2000] (DSPE-PEG2000). Microbubble size and concentration were measured as a function of time and temperature by optical microscopy. Effects on MG63 osteosarcoma and human bone marrow stromal cells (BMSCs) were measured for up to 72 h by assay for viability, metabolic activity and proliferation. RESULTS DBPC:DSPE-PEG2000 microbubbles were significantly more stable than DSPC:PEG40S microbubbles under all conditions tested. Serum-containing medium had no detrimental effect on microbubble stability, but storage at 37 °C compared to at 4 °C reduced stability for both preparations, with almost complete dissolution of microbubbles at times ≥24 h. DSPC:PEG40S microbubbles had greater inhibitory effects on cell metabolism and growth than DBPC:DSPE-PEG2000 microbubbles, with PEG40S found to be the principle inhibitory component. These effects were only evident at high microbubble concentrations (≥20% (v/v)) or with prolonged culture (≥24 h). Increasing cell-microbubble contact by inversion culture in a custom-built device had no inhibitory effect on metabolism. CONCLUSIONS These data indicate that, over a broad range of concentrations and incubation times, DBPC:DSPE-PEG2000 and DSPC:PEG40S microbubbles have little effect on osteoblastic cell viability and growth, and that PEG40S is the principle inhibitory component in the formulations investigated.
Collapse
Affiliation(s)
- A E Polydorou
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research group, University of Southampton, United Kingdom; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom
| | - J P May
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research group, University of Southampton, United Kingdom; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom
| | - K Makris
- Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom
| | - S Ferri
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research group, University of Southampton, United Kingdom; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom
| | - Q Wu
- Institute of Biomedical Engineering, University of Oxford, United Kingdom
| | - E Stride
- Institute of Biomedical Engineering, University of Oxford, United Kingdom; Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, United Kingdom
| | - D Carugo
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, United Kingdom
| | - N D Evans
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research group, University of Southampton, United Kingdom; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom.
| |
Collapse
|
6
|
Jang S, Bustos SS, Chen AD, Zheng EE, Hesley GK, Brinkman NJ, Carter JS, Tran NV, Fahradyan V, Lee CU. Lymphatic Mapping with Contrast-enhanced Ultrasound for Lymphaticovenous Anastomosis Surgery: How We Do It. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2023; 11:e5328. [PMID: 37842077 PMCID: PMC10569760 DOI: 10.1097/gox.0000000000005328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 08/31/2023] [Indexed: 10/17/2023]
Abstract
Lymphaticovenous anastomosis (LVA) surgery is an effective surgery for the treatment of lymphedema in the extremities. Indocyanine green lymphography is the reference standard for visualizing lymphatics for LVA surgery, but it has several limitations; most notably, superficial dermal congestion can mask deeper lymphatic vessels. To overcome the limitations, we add contrast-enhanced ultrasound (CEUS) lymphography. We have previously reported that CEUS lymphography can identify lymphatic vessels for LVA surgery that indocyanine green lymphography does not. Here, we describe how we perform CEUS lymphography, including workflow, technique, and documentation. Before informed consent, the patient must be screened for possible adverse reactions to microbubbles. The procedure involves multiple intradermal injections of the microbubble agent at various sites along the extremity. After each injection, imaging for microbubble uptake by lymphatic vessels is performed using an ultrasound scanner with contrast-specific software. We use sulfur hexafluoride lipid-type A microspheres (Lumason/SonoVue; Bracco Suisse SA), but we are investigating the performance of other Food & Drug Administration-approved microbubble agents for CEUS lymphography. Having a systematic approach to marking the skin can mitigate the hindrance of marking over ultrasound coupling gel. Another benefit of CEUS lymphography is the rapid identification of neighboring veins compatible in size and location for anastomosis. We hold regular scheduled multidisciplinary meetings for coordination of care, discussion of outcomes, quality assurance, and ongoing innovation.
Collapse
Affiliation(s)
- Samuel Jang
- From the Department of Radiology, Mayo Clinic, Rochester, Minn
| | | | - Austin D. Chen
- Department of Plastic Surgery, Mayo Clinic, Rochester, Minn
| | | | - Gina K. Hesley
- From the Department of Radiology, Mayo Clinic, Rochester, Minn
| | | | - Jill S. Carter
- From the Department of Radiology, Mayo Clinic, Rochester, Minn
| | - Nho V. Tran
- Department of Plastic Surgery, Mayo Clinic, Rochester, Minn
| | - Vahe Fahradyan
- Department of Plastic Surgery, Mayo Clinic, Rochester, Minn
| | | |
Collapse
|
7
|
Mee-Inta O, Hsieh CF, Chen DQ, Fan CH, Chiang YY, Liu CC, Sze CI, Gean PW, Wu PC, Yang MS, Huang PS, Chieh Wu P, Kuo YM, Huang CC. High-frequency ultrasound imaging for monitoring the function of meningeal lymphatic system in mice. ULTRASONICS 2023; 131:106949. [PMID: 36773481 DOI: 10.1016/j.ultras.2023.106949] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/30/2022] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
The meningeal lymphatic system drains the cerebrospinal fluid from the subarachnoid space to the cervical lymphatic system, primarily to the deep cervical lymph nodes. Perturbations of the meningeal lymphatic system have been linked to various neurologic disorders. A method to specifically monitor the flow of meningeal lymphatic system in real time is unavailable. In the present study, we adopted the high-frequency ultrasound (HFUS) with 1,1'diocatadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI)-loaded microbubble and FePt@PLGA nanoparticle contrast agents to evaluate the flow of the meningeal lymphatic system in 2-month-old mice. Statistical analysis was performed to identify changes of HFUS signals among the microbubbles, FePt@PLGA nanoparticles, and saline control groups. Approximately 15 min from the start of intracerebroventricular injection of contrast agents, their signals were evident at the deep cervical lymph nodes and lasted for at least 60 min. These signals were validated on the basis of the presence of DiI and Fe signals in the deep cervical lymph nodes. Ligation of afferent lymphatic vessels to the deep cervical lymph nodes eliminated the HFUS signals. Moreover, ablation of lymphatic vessels near the confluence of sinuses decreased the HFUS signals in the deep cervical lymph nodes. Glioma-bearing mice that exhibited reduced lymphatic vessel immunostaining signals near the confluence of sinuses had lowered HFUS signals in the deep cervical lymph nodes within 60 min. The proposed method provides a minimally invasive approach to monitor the qualities of the meningeal lymphatic system in real time as well as the progression of the meningeal lymphatic system in various brain disease animal models.
Collapse
Affiliation(s)
- Onanong Mee-Inta
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chin-Fang Hsieh
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - De-Quan Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Yi Chiang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Chan-Chuan Liu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Chun-I Sze
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Wu Gean
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ping-Ching Wu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan; Institute of Oral Medicine and Department of Stomatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University Tainan, Taiwan; Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| | - Mon-Shieh Yang
- College of Science, National Cheng Kung University, Tainan, Taiwan
| | - Po-Sheng Huang
- Department of Photonics, National Cheng Kung University, Tainan, Taiwan
| | - Pin Chieh Wu
- Department of Photonics, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Chih-Chung Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
8
|
Barbagianni MS, Gouletsou PG. Modern Imaging Techniques in the Study and Disease Diagnosis of the Mammary Glands of Animals. Vet Sci 2023; 10:vetsci10020083. [PMID: 36851387 PMCID: PMC9965774 DOI: 10.3390/vetsci10020083] [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: 12/07/2022] [Revised: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
The study of the structure and function of the animals' mammary glands is of key importance, as it reveals pathological processes at their onset, thus contributing to their immediate treatment. The most frequently studied mammary diseases are mastitis in cows and ewes and mammary tumours in dogs and cats. Various imaging techniques such as computed tomography, positron emission tomography, magnetic resonance imaging, and ultrasonographic techniques (Doppler, contrast-enchanced, three-dimensional and elastography) are available and can be applied in research or clinical practice in order to evaluate possible abnormalities in mammary glands, as well as to assist in the differential diagnosis. In this review, the above imaging technologies are described, and the perspectives of each method are highlighted. It is inferred that ultrasonographic modalities are the most frequently used imaging techniques for the diagnosis of clinical or subclinical mastitis and treatment guidance on a farm. In companion animals, a combination of imaging techniques should be applied for a more accurate diagnosis of mammary tumours. In any case, the confirmation of the diagnosis is provided by laboratory techniques.
Collapse
|
9
|
Leow KS, Kwok CY, Low HM, Lohan R, Lim TC, Low SCA, Tan CH. Algorithm‐based approach to focal liver lesions in contrast‐enhanced ultrasound. Australas J Ultrasound Med 2022; 25:142-153. [DOI: 10.1002/ajum.12306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Kheng Song Leow
- Department of Radiology Woodlands Health Campus 2 Yishun Central 2, Tower E, Level 5 Singapore Singapore
| | - Christine Ying Kwok
- Department of Diagnostic Radiology Tan Tock Seng Hospital 11 Jalan Tan Tock Seng Singapore 308433 Singapore
| | - Hsien Min Low
- Department of Diagnostic Radiology Tan Tock Seng Hospital 11 Jalan Tan Tock Seng Singapore 308433 Singapore
| | - Rahul Lohan
- Department of Diagnostic Radiology Khoo Teck Puat Hospital 90 Yishun Central Singapore Singapore
| | - Tze Chwan Lim
- Department of Radiology Woodlands Health Campus 2 Yishun Central 2, Tower E, Level 5 Singapore Singapore
| | - Su Chong Albert Low
- Department of Diagnostic Radiology Singapore General Hospital Outram Road Singapore 169608 Singapore
| | - Cher Heng Tan
- Department of Diagnostic Radiology Tan Tock Seng Hospital 11 Jalan Tan Tock Seng Singapore 308433 Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| |
Collapse
|
10
|
Carlomagno F, Pozza C, Tenuta M, Pofi R, Tarani L, Sesti F, Minnetti M, Gianfrilli D, Isidori AM. Testicular Microvascular Flow Is Altered in Klinefelter Syndrome and Predicts Circulating Testosterone. J Clin Endocrinol Metab 2022; 107:e236-e245. [PMID: 34407199 DOI: 10.1210/clinem/dgab605] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Indexed: 01/20/2023]
Abstract
CONTEXT Experimental studies on Klinefelter syndrome (KS) reported increased intratesticular testosterone (T) levels coexisting with reduced circulating levels. Abnormalities in testicular microcirculation have been claimed; however, no studies investigated in vivo testicular blood flow dynamics in humans with KS. OBJECTIVE To analyze the testicular microcirculation in KS by contrast-enhanced ultrasonography (CEUS) and correlate vascular parameters with endocrine function. DESIGN AND SETTING Prospective study. University setting. PATIENTS Sixty-eight testicular scans, 34 testes from 19 T-naïve subjects with KS and 34 testes from age-matched eugonadal men (control) who underwent CEUS for incidental nonpalpable testicular lesions. MAIN OUTCOMES CEUS kinetic parameters. RESULTS CEUS revealed slower testicular perfusion kinetics in subjects with KS than in age-matched controls. Specifically, the wash-in time (P = 0.018), mean transit time (P = 0.035), time to peak (P < 0.001), and wash-out time (P = 0.004) were all prolonged. Faster testicular blood flow was associated with higher total T levels. Principal component analysis and multiple linear regression analyses confirmed the findings and supported a role for reduced venous blood flow as independent predictor of total T levels. CONCLUSIONS Testicular venous blood flow is altered in KS and independently predicts T peripheral release.
Collapse
Affiliation(s)
- Francesco Carlomagno
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Carlotta Pozza
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Marta Tenuta
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Luigi Tarani
- Department of Pediatrics, Sapienza University of Rome, 00161, Rome, Italy
| | - Franz Sesti
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Marianna Minnetti
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| |
Collapse
|
11
|
Hai Y, Savsani E, Chong W, Eisenbrey J, Lyshchik A. Meta-analysis and systematic review of contrast-enhanced ultrasound in evaluating the treatment response after locoregional therapy of hepatocellular carcinoma. Abdom Radiol (NY) 2021; 46:5162-5179. [PMID: 34410432 DOI: 10.1007/s00261-021-03248-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Contrast-enhanced ultrasound (CEUS) is a useful tool to assess treatment response after percutaneous ablation or transarterial chemoembolization (TACE) of hepatocellular carcinoma (HCC). Here, we performed a systematic review and meta-analysis to evaluate the usefulness of CEUS in identifying residual tumor after locoregional therapy. METHODS PubMed, Scopus, and Cochrane library databases were searched from their inception until March 8, 2021, for diagnostic test accuracy studies comparing CEUS to a reference standard for identifying residual tumors after locoregional therapy of HCC. The pooled sensitivity, specificity, accuracy, and diagnostic odds ratio (DOR) were obtained using a bivariate random effects model. Subgroup analyses were performed by stratifying the studies based on study design, type of locoregional therapy, CEUS criteria for residual tumor, timing of CEUS follow up, and type of standard reference. RESULTS Two reviewers independently evaluated 1479 publications. After full-text review, 142 studies were found to be relevant, and 43 publications (50 cohorts) were finally included. The overall sensitivity of CEUS in detection of residual disease estimated from the bivariate random effects model was 0.85 (95% CI 0.80-0.89). Similarly, the overall specificity was 0.94 (95% CI 0.91-0.96). The diagnostic accuracy was 93.5%. The DOR was 70.1 (95% CI 62.2-148), and the AUROC was 0.95. Importantly, subgroup analysis showed no apparent differences in the diagnostic performance between locoregional therapy (TACE vs. ablation) and criteria used to define residual enhancement, timing of performing CEUS, study design, or type of reference standard. CONCLUSION CEUS is a highly accurate method to identify HCC residual tumor after TACE or percutaneous ablation.
Collapse
Affiliation(s)
- Yang Hai
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, 763G Main Building, Philadelphia, PA, 19107, USA
| | - Esika Savsani
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, 763G Main Building, Philadelphia, PA, 19107, USA
| | - Weelic Chong
- Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut St, Suite 100, Philadelphia, PA, USA
- Department of Medical Oncology, Thomas Jefferson University, 1025 Walnut St, Suite 727, Philadelphia, PA, USA
| | - John Eisenbrey
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, 763G Main Building, Philadelphia, PA, 19107, USA
| | - Andrej Lyshchik
- Department of Radiology, Thomas Jefferson University, 132 South 10th Street, 763G Main Building, Philadelphia, PA, 19107, USA.
| |
Collapse
|
12
|
Krafft MP, Riess JG. Therapeutic oxygen delivery by perfluorocarbon-based colloids. Adv Colloid Interface Sci 2021; 294:102407. [PMID: 34120037 DOI: 10.1016/j.cis.2021.102407] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
After the protocol-related indecisive clinical trial of Oxygent, a perfluorooctylbromide/phospholipid nanoemulsion, in cardiac surgery, that often unduly assigned the observed untoward effects to the product, the development of perfluorocarbon (PFC)-based O2 nanoemulsions ("blood substitutes") has come to a low. Yet, significant further demonstrations of PFC O2-delivery efficacy have continuously been reported, such as relief of hypoxia after myocardial infarction or stroke; protection of vital organs during surgery; potentiation of O2-dependent cancer therapies, including radio-, photodynamic-, chemo- and immunotherapies; regeneration of damaged nerve, bone or cartilage; preservation of organ grafts destined for transplantation; and control of gas supply in tissue engineering and biotechnological productions. PFC colloids capable of augmenting O2 delivery include primarily injectable PFC nanoemulsions, microbubbles and phase-shift nanoemulsions. Careful selection of PFC and other colloid components is critical. The basics of O2 delivery by PFC nanoemulsions will be briefly reminded. Improved knowledge of O2 delivery mechanisms has been acquired. Advanced, size-adjustable O2-delivering nanoemulsions have been designed that have extended room-temperature shelf-stability. Alternate O2 delivery options are being investigated that rely on injectable PFC-stabilized microbubbles or phase-shift PFC nanoemulsions. The latter combine prolonged circulation in the vasculature, capacity for penetrating tumor tissues, and acute responsiveness to ultrasound and other external stimuli. Progress in microbubble and phase-shift emulsion engineering, control of phase-shift activation (vaporization), understanding and control of bubble/ultrasound/tissue interactions is discussed. Control of the phase-shift event and of microbubble size require utmost attention. Further PFC-based colloidal systems, including polymeric micelles, PFC-loaded organic or inorganic nanoparticles and scaffolds, have been devised that also carry substantial amounts of O2. Local, on-demand O2 delivery can be triggered by external stimuli, including focused ultrasound irradiation or tumor microenvironment. PFC colloid functionalization and targeting can help adjust their properties for specific indications, augment their efficacy, improve safety profiles, and expand the range of their indications. Many new medical and biotechnological applications involving fluorinated colloids are being assessed, including in the clinic. Further uses of PFC-based colloidal nanotherapeutics will be briefly mentioned that concern contrast diagnostic imaging, including molecular imaging and immune cell tracking; controlled delivery of therapeutic energy, as for noninvasive surgical ablation and sonothrombolysis; and delivery of drugs and genes, including across the blood-brain barrier. Even when the fluorinated colloids investigated are designed for other purposes than O2 supply, they will inevitably also carry and deliver a certain amount of O2, and may thus be considered for O2 delivery or co-delivery applications. Conversely, O2-carrying PFC nanoemulsions possess by nature a unique aptitude for 19F MR imaging, and hence, cell tracking, while PFC-stabilized microbubbles are ideal resonators for ultrasound contrast imaging and can undergo precise manipulation and on-demand destruction by ultrasound waves, thereby opening multiple theranostic opportunities.
Collapse
Affiliation(s)
- Marie Pierre Krafft
- University of Strasbourg, Institut Charles Sadron (CNRS), 23 rue du Loess, 67034 Strasbourg, France.
| | - Jean G Riess
- Harangoutte Institute, 68160 Ste Croix-aux-Mines, France
| |
Collapse
|
13
|
Abstract
Contrast-enhanced ultrasound (CEUS) is a safe adjunct tool for liver imaging and can be an alternative to computed tomography or MR imaging. CEUS has a proven track record in guiding management for patients with chronic liver disease who need further evaluation of focal liver lesions. CEUS is a dynamic examination with high temporal and spatial resolution. CEUS uses a pure blood pool contrast agent that allows for a unique evaluation of the perfusion kinetics of a region of interest.
Collapse
Affiliation(s)
- Stephanie Spann
- Department of Radiology, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, E6-230-BF, Dallas, TX 75390-9316, USA
| | - David T Fetzer
- Department of Radiology, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, E6-230-BF, Dallas, TX 75390-9316, USA.
| |
Collapse
|
14
|
Baranger J, Villemain O, Wagner M, Vargas-Gutierrez M, Seed M, Baud O, Ertl-Wagner B, Aguet J. Brain perfusion imaging in neonates. NEUROIMAGE-CLINICAL 2021; 31:102756. [PMID: 34298475 PMCID: PMC8319803 DOI: 10.1016/j.nicl.2021.102756] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 06/21/2021] [Accepted: 07/03/2021] [Indexed: 02/07/2023]
Abstract
MRI is the modality of choice to image and quantify cerebral perfusion. Imaging of neonatal brain perfusion is possible using MRI and ultrasound. Novel ultrafast ultrasound imaging allows for excellent spatiotemporal resolution. Understanding cerebral hemodynamic changes of neonatal adaptation is key.
Abnormal variations of the neonatal brain perfusion can result in long-term neurodevelopmental consequences and cerebral perfusion imaging can play an important role in diagnostic and therapeutic decision-making. To identify at-risk situations, perfusion imaging of the neonatal brain must accurately evaluate both regional and global perfusion. To date, neonatal cerebral perfusion assessment remains challenging. The available modalities such as magnetic resonance imaging (MRI), ultrasound imaging, computed tomography (CT), near-infrared spectroscopy or nuclear imaging have multiple compromises and limitations. Several promising methods are being developed to achieve better diagnostic accuracy and higher robustness, in particular using advanced MRI and ultrasound techniques. The objective of this state-of-the-art review is to analyze the methodology and challenges of neonatal brain perfusion imaging, to describe the currently available modalities, and to outline future perspectives.
Collapse
Affiliation(s)
- Jérôme Baranger
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Translation Medicine Department, SickKids Research Institute, Toronto, Ontario, Canada
| | - Olivier Villemain
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Translation Medicine Department, SickKids Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Matthias Wagner
- Department of Diagnostic Imaging, Division of Neuroradiology, The Hospital for Sick Children, Toronto, Canada
| | | | - Mike Seed
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Translation Medicine Department, SickKids Research Institute, Toronto, Ontario, Canada; Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada
| | - Olivier Baud
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva and University of Geneva, Geneva, Switzerland
| | - Birgit Ertl-Wagner
- Department of Diagnostic Imaging, Division of Neuroradiology, The Hospital for Sick Children, Toronto, Canada
| | - Julien Aguet
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada.
| |
Collapse
|
15
|
Pellow C, Cherin E, Abenojar EC, Exner AA, Zheng G, Demore CEM, Goertz DE. High-Frequency Array-Based Nanobubble Nonlinear Imaging in a Phantom and In Vivo. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:2059-2074. [PMID: 33513102 PMCID: PMC8296974 DOI: 10.1109/tuffc.2021.3055141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
There has been growing interest in nanobubbles (NBs) for vascular and extravascular ultrasound contrast imaging and therapeutic applications. Studies to date have generally utilized low frequencies (<12 MHz), high concentrations (>109 mL-1), and uncalibrated B-mode or contrast-mode on commercial systems without reporting investigations on NB signatures upon which the imaging protocols should be based. We recently demonstrated that low concentrations (106 mL-1) of porphyrin-lipid-encapsulated NBs scatter nonlinearly at low (2.5, 8 MHz) and high (12.5, 25, 30 MHz) frequencies in a pressure threshold-dependent manner that is advantageous for amplitude modulation (AM) imaging. Here, we implement pressure-calibrated AM at high frequency on a commercial preclinical array system to enhance sensitivity to nonlinear scattering of three phospholipid-based NB formulations. With this approach, improvements in contrast to tissue ratio relative to B-mode between 12.4 and 22.8 dB are demonstrated in a tissue-mimicking phantom, and between 6.7 and 14.8 dB in vivo.
Collapse
|
16
|
Tenuta M, Sesti F, Bonaventura I, Mazzotta P, Pofi R, Gianfrilli D, Pozza C. Use of contrast enhanced ultrasound in testicular diseases: A comprehensive review. Andrology 2021; 9:1369-1382. [PMID: 34043256 PMCID: PMC8640938 DOI: 10.1111/andr.13057] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 01/17/2023]
Abstract
Background Contrast‐enhanced ultrasound (CEUS) is a sonographic technique that increases the diagnostic accuracy of ultrasound and color Doppler ultrasound (CDUS) when studying testicular abnormalities. However, its role in clinical practice is still debatable because there are no accepted standards regarding how and when this technique should be used for patients with testicular disease. Objectives To perform a nonsystematic review of the current literature to highlight the strength and flaws of performing CEUS and to provide a critical overview of current research evidence on this topic. Materials and methods A thorough search of published peer‐reviewed studies in PubMed was performed using proper keywords. Results Strong enhancement of neoplastic lesions (both benign and malignant) during CEUS aids in differential diagnosis with non‐neoplastic lesions, which usually appears either nonenhanced or enhanced in a manner similar to that of the surrounding parenchyma. CEUS enhancement has a high predictive value in the identification of neoplastic lesions, whereas a similar or complete absence of enhancement may be interpreted as strong evidence of benignity, although there are exceptions. Literature on quantitative analysis is still scarce, though promising, particularly in distinguishing benign from malignant neoplasms. Furthermore, CEUS may be useful in many emergency situations, such as acute scrotum, blunt scrotal trauma, and focal infarction of the testis. Finally, CEUS can help increase the probability of sperm recovery in azoospermic males. Discussion and conclusion CEUS is a safe, easy‐to‐perform, and cost‐effective diagnostic tool that can provide a more accurate diagnosis in testicular lesions and acute scrotal disease. However, further studies with larger cohorts are required to refine the differential diagnosis between benign and malignant neoplasms. Finally, these preliminary results can instigate the development of innovative research on pre‐testicular sperm extraction to increase the chances of sperm recovery.
Collapse
Affiliation(s)
- Marta Tenuta
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Franz Sesti
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Paola Mazzotta
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Carlotta Pozza
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
17
|
Lafond M, Salido NG, Haworth KJ, Hannah AS, Macke GP, Genstler C, Holland CK. Cavitation Emissions Nucleated by Definity Infused through an EkoSonic Catheter in a Flow Phantom. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:693-709. [PMID: 33349516 DOI: 10.1016/j.ultrasmedbio.2020.10.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/05/2020] [Accepted: 10/18/2020] [Indexed: 06/12/2023]
Abstract
The EkoSonic endovascular system has been cleared by the U.S. Food and Drug Administration for the controlled and selective infusion of physician specified fluids, including thrombolytics, into the peripheral vasculature and the pulmonary arteries. The objective of this study was to explore whether this catheter technology could sustain cavitation nucleated by infused Definity, to support subsequent studies of ultrasound-mediated drug delivery to diseased arteries. The concentration and attenuation spectroscopy of Definity were assayed before and after infusion at 0.3, 2.0 and 4.0 mL/min through the EkoSonic catheter. PCI was used to map and quantify stable and inertial cavitation as a function of Definity concentration in a flow phantom mimicking the porcine femoral artery. The 2.0 mL/min infusion rate yielded the highest surviving Definity concentration and acoustic attenuation. Cavitation was sustained throughout each 15 ms ultrasound pulse, as well as throughout the 3 min infusion. These results demonstrate a potential pathway to use cavitation nucleation to promote drug delivery with the EkoSonic endovascular system.
Collapse
Affiliation(s)
- Maxime Lafond
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA.
| | - Nuria G Salido
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kevin J Haworth
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Gregory P Macke
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Christy K Holland
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
18
|
Zhang C, Li Y, Ma X, He W, Liu C, Liu Z. Functional micro/nanobubbles for ultrasound medicine and visualizable guidance. Sci China Chem 2021; 64:899-914. [PMID: 33679901 PMCID: PMC7921288 DOI: 10.1007/s11426-020-9945-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/18/2021] [Indexed: 12/28/2022]
Abstract
Chemically functionalized gas-filled bubbles with a versatile micro/nano-sized scale have witnessed a long history of developments and emerging applications in disease diagnosis and treatments. In combination with ultrasound and image-guidance, micro/nanobubbles have been endowed with the capabilities of biomedical imaging, drug delivery, gene transfection and disease-oriented therapy. As an external stimulus, ultrasound (US)-mediated targeting treatments have been achieving unprecedented efficiency. Nowadays, US is playing a crucial role in visualizing biological/pathological changes in lives as a reliable imaging technique and a powerful therapeutic tool. This review retrospects the history of ultrasound, the chemistry of functionalized agents and summarizes recent advancements of functional micro/nanobubbles as US contrast agents in preclinical and transclinical research. Latest ultrasound-based treatment modalities in association with functional micro/nanobubbles have been highlighted as their great potentials for disease precision therapy. It is believed that these state-of-the-art micro/nanobubbles will become a booster for ultrasound medicine and visualizable guidance to serve future human healthcare in a more comprehensive and practical manner.
Collapse
Affiliation(s)
- Chen Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin, 300072 China
| | - Yihong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin, 300072 China
| | - Xinyong Ma
- Division of Academic & Cultural Activities, Academic Divisions of the Chinese Academy of Sciences, Beijing, 100190 China
| | - Wenxin He
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin, 300072 China
| | - Chenxi Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin, 300072 China
| | - Zhe Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin, 300072 China
| |
Collapse
|
19
|
Krishnan K, Bhutani MS, Aslanian HR, Melson J, Navaneethan U, Pannala R, Parsi MA, Schulman AR, Sethi A, Sullivan S, Trikudanathan G, Trindade AJ, Watson RR, Maple JT, Lichtenstein DR. Enhanced EUS imaging (with videos). Gastrointest Endosc 2021; 93:323-333. [PMID: 33129492 DOI: 10.1016/j.gie.2020.06.075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS EUS remains a primary diagnostic tool for the evaluation of pancreaticobiliary disease. Although EUS combined with FNA or biopsy sampling is highly sensitive for the diagnosis of neoplasia within the pancreaticobiliary tract, limitations exist in specific clinical settings such as chronic pancreatitis. Enhanced EUS imaging technologies aim to aid in the detection and diagnosis of lesions that are commonly evaluated with EUS. METHODS We reviewed technologies and methods for enhanced imaging during EUS and applications of these methods. Available data regarding efficacy, safety, and financial considerations are summarized. RESULTS Enhanced EUS imaging methods include elastography and contrast-enhanced EUS (CE-EUS). Both technologies have been best studied in the setting of pancreatic mass lesions. Robust data indicate that neither technology has adequate specificity to serve as a stand-alone test for pancreatic malignancy. However, there may be a role for improving the targeting of sampling and in the evaluation of peritumoral lymph nodes, inflammatory pancreatic masses, and masses with nondiagnostic FNA or fine-needle biopsy sampling. Further, novel applications of these technologies have been reported in the evaluation of liver fibrosis, pancreatic cysts, and angiogenesis within neoplastic lesions. CONCLUSIONS Elastography and CE-EUS may improve the real-time evaluation of intra- and extraluminal lesions as an adjunct to standard B-mode and Doppler imaging. They are not a replacement for EUS-guided tissue sampling but provide adjunctive diagnostic information in specific clinical situations. The optimal clinical use of these technologies continues to be a focus of ongoing research.
Collapse
Affiliation(s)
- Kumar Krishnan
- Division of Gastroenterology, Department of Internal Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Manoop S Bhutani
- Department of Gastroenterology Hepatology and Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Harry R Aslanian
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joshua Melson
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | | | - Rahul Pannala
- Department of Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Mansour A Parsi
- Section for Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Allison R Schulman
- Department of Gastroenterology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Amrita Sethi
- New York-Presbyterian Medical Center/Columbia University Medical Center, New York, New York, USA
| | - Shelby Sullivan
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Guru Trikudanathan
- Department of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Arvind J Trindade
- Department of Gastroenterology, Zucker School of Medicine at Hofstra/Northwell, Long Island Jewish Medical Center, New Hyde Park, New York, USA
| | - Rabindra R Watson
- Department of Gastroenterology, Interventional Endoscopy Services, California Pacific Medical Center, San Francisco, California, USA
| | - John T Maple
- Division of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - David R Lichtenstein
- Division of Gastroenterology, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
20
|
Notohamiprodjo S, Treitl KM, Hauke C, Sutter SM, Auweter S, Pfeiffer F, Reiser MF, Hellbach K. Imaging characteristics of intravascular spherical contrast agents for grating-based x-ray dark-field imaging – effects of concentrations, spherical sizes and applied voltage. Sci Rep 2020; 10:9405. [PMID: 32523085 PMCID: PMC7287139 DOI: 10.1038/s41598-020-66395-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/14/2020] [Indexed: 11/09/2022] Open
Abstract
AbstractThis study investigates the x-ray scattering characteristics of microsphere particles in x-ray-grating-based interferometric imaging at different concentrations, bubble sizes and tube voltages (kV). Attenuation (ATI), dark-field (DFI) and phase-contrast (PCI) images were acquired. Signal-to-noise (SNR) and contrast-to-noise ratios with water (CNRw) and air as reference (CNRa) were determined. In all modalities, a linear relationship between SNR and microbubbles concentration, respectively, microsphere size was found. A significant gain of SNR was found when varying kV. SNR was significantly higher in DFI and PCI than ATI. The highest gain of SNR was shown at 60 kV for all media in ATI and DFI, at 80 kV for PCI. SNR for all media was significantly higher compared to air and was slightly lower compared to water. A linear relationship was found between CNRa, CNRw, concentration and size. With increasing concentration and decreasing size, CNRa and CNRw increased in DFI, but decreased in PCI. Best CNRa and CNRw was found at specific combination of kV and concentration/size. Highest average CNRa and CNRw was found for microspheres in ATI and PCI, for microbubbles in DFI. Microspheres are a promising contrast-media for grating-based-interferometry, if kV, microsphere size and concentration are appropriately combined.
Collapse
|
21
|
Bam R, Daryaei I, Abou-Elkacem L, Vilches-Moure JG, Meuillet EJ, Lutz A, Marinelli ER, Unger EC, Gambhir SS, Paulmurugan R. Toward the Clinical Development and Validation of a Thy1-Targeted Ultrasound Contrast Agent for the Early Detection of Pancreatic Ductal Adenocarcinoma. Invest Radiol 2020; 55:711-721. [PMID: 32569010 PMCID: PMC7541735 DOI: 10.1097/rli.0000000000000697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Early detection of pancreatic ductal adenocarcinoma (PDAC) represents the most significant step toward the treatment of this aggressive lethal disease. Previously, we engineered a preclinical Thy1-targeted microbubble (MBThy1) contrast agent that specifically recognizes Thy1 antigen overexpressed in the vasculature of murine PDAC tissues by ultrasound (US) imaging. In this study, we adopted a single-chain variable fragment (scFv) site-specific bioconjugation approach to construct clinically translatable MBThy1-scFv and test for its efficacy in vivo in murine PDAC imaging, and functionally evaluated the binding specificity of scFv ligand to human Thy1 in patient PDAC tissues ex vivo. MATERIALS AND METHODS We recombinantly expressed the Thy1-scFv with a carboxy-terminus cysteine residue to facilitate its thioether conjugation to the PEGylated MBs presenting with maleimide functional groups. After the scFv-MB conjugations, we tested binding activity of the MBThy1-scFv to MS1 cells overexpressing human Thy1 (MS1Thy1) under liquid shear stress conditions in vitro using a flow chamber setup at 0.6 mL/min flow rate, corresponding to a wall shear stress rate of 100 seconds, similar to that in tumor capillaries. For in vivo Thy1 US molecular imaging, MBThy1-scFv was tested in the transgenic mouse model (C57BL/6J - Pdx1-Cre; KRas; Ink4a/Arf) of PDAC and in control mice (C57BL/6J) with L-arginine-induced pancreatitis or normal pancreas. To facilitate its clinical feasibility, we further produced Thy1-scFv without the bacterial fusion tags and confirmed its recognition of human Thy1 in cell lines by flow cytometry and in patient PDAC frozen tissue sections of different clinical grades by immunofluorescence staining. RESULTS Under shear stress flow conditions in vitro, MBThy1-scFv bound to MS1Thy1 cells at significantly higher numbers (3.0 ± 0.8 MB/cell; P < 0.01) compared with MBNontargeted (0.5 ± 0.5 MB/cell). In vivo, MBThy1-scFv (5.3 ± 1.9 arbitrary units [a.u.]) but not the MBNontargeted (1.2 ± 1.0 a.u.) produced high US molecular imaging signal (4.4-fold vs MBNontargeted; n = 8; P < 0.01) in the transgenic mice with spontaneous PDAC tumors (2-6 mm). Imaging signal from mice with L-arginine-induced pancreatitis (n = 8) or normal pancreas (n = 3) were not significantly different between the two MB constructs and were significantly lower than PDAC Thy1 molecular signal. Clinical-grade scFv conjugated to Alexa Fluor 647 dye recognized MS1Thy1 cells but not the parental wild-type cells as evaluated by flow cytometry. More importantly, scFv showed highly specific binding to VEGFR2-positive vasculature and fibroblast-like stromal components surrounding the ducts of human PDAC tissues as evaluated by confocal microscopy. CONCLUSIONS Our findings summarize the development and validation of a clinically relevant Thy1-targeted US contrast agent for the early detection of human PDAC by US molecular imaging.
Collapse
Affiliation(s)
- Rakesh Bam
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| | | | - Lotfi Abou-Elkacem
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| | | | | | - Amelie Lutz
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| | | | | | - Sanjiv S. Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| |
Collapse
|
22
|
Karagozoglu KH, Helder M, Bot J, Kamp O, Forouzanfar T, Brand HS, Cha S, Weisman G, Bartelink I, Vissink A, Jager DHJ. Intraoperative visualisation and treatment of salivary glands in Sjögren's syndrome by contrast-enhanced ultrasound sialendoscopy (CEUSS): protocol for a phase I single-centre, single-arm, exploratory study. BMJ Open 2020; 10:e033542. [PMID: 32998913 PMCID: PMC7528357 DOI: 10.1136/bmjopen-2019-033542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION We established a promising sialendoscopic treatment for in vivo enhancement of salivation in salivary glands affected by Sjögren's syndrome (SS). In this technique, the ducts of the salivary glands are irrigated with saline and steroids. This allows for dilatation of ductal strictures and removal of debris. Unfortunately, it is not possible to assess the delivery and penetration of saline or medications in the ductal system and parenchyma. To address this problem, we will conduct contrast-enhanced ultrasound sialendoscopy (CEUSS) using sulphur hexafluoride microbubbles. To the best of our knowledge, microbubbles have never been used for the treatment of salivary glands in SS. It is, therefore, imperative to test this application for its safety and feasibility. METHODS AND ANALYSIS A single-arm phase I study will be performed in 10 SS patients. Under local anaesthesia, ultrasound (US) guided infusion of the parotid and submandibular glands with microbubbles will be performed. Continuous US imaging will be used to visualise the glands, including the location of strictures and occlusions. Main outcomes will be the evaluation of safety and technical feasibility of the experimental treatment. Secondary outcomes will consist of determinations of unstimulated whole mouth saliva flow, stimulated whole mouth saliva flow, stimulated parotid saliva flow, clinical oral dryness, reported pain, xerostomia, disease activity, salivary cytokine profiles and clinical SS symptoms. Finally, salivary gland topographical alterations will be evaluated by US. ETHICS AND DISSEMINATION Ethical approval for this study was obtained from the Medical Ethics Committee of the Amsterdam University Medical Centre, Amsterdam, The Netherlands (NL68283.029.19). data will be presented at national and international conferences and published in a peer-reviewed journal. The study will be implemented and reported in line with the Standard Protocol Items: Recommendations for Interventional Trials' statement. TRIAL REGISTRATION NUMBERS The Netherlands Trial Register: NL7731, MREC Trial Register: NL68283.029.19; Pre-results.
Collapse
Affiliation(s)
- K Hakki Karagozoglu
- Department of Oral and Maxillofacial Surgery / Oral Pathology, Amsterdam University Medical Center (Amsterdam UMC, Location VUmc) and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marco Helder
- Department of Oral and Maxillofacial Surgery / Oral Pathology, Amsterdam University Medical Center (Amsterdam UMC, Location VUmc) and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Joseph Bot
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center (Amsterdam UMC, Location VUmc), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Otto Kamp
- Department of Cardiology, Amsterdam University Medical Center (Amsterdam UMC, Location VUmc), Amsterdam, The Netherlands
| | - Tim Forouzanfar
- Department of Oral and Maxillofacial Surgery / Oral Pathology, Amsterdam University Medical Center (Amsterdam UMC, Location VUmc) and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Henk S Brand
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | - Seunghee Cha
- Department of Oral and Maxillofacial Diagnostic Sciences, Division of Oral Medicine, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Gary Weisman
- Department of Biochemistry, MU Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Imke Bartelink
- Department of Clinical Pharmacology and Pharmacy, Amsterdam University Medical Center (Amsterdam UMC, Location VUmc), Amsterdam, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Derk Hendrik Jan Jager
- Department of Oral and Maxillofacial Surgery / Oral Pathology, Amsterdam University Medical Center (Amsterdam UMC, Location VUmc) and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Pellow C, Abenojar EC, Exner AA, Zheng G, Goertz DE. Concurrent visual and acoustic tracking of passive and active delivery of nanobubbles to tumors. Am J Cancer Res 2020; 10:11690-11706. [PMID: 33052241 PMCID: PMC7545999 DOI: 10.7150/thno.51316] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Background: There has been growing interest in nanobubbles for their potential to extend bubble-mediated ultrasound approaches beyond that of their larger microbubble counterparts. In particular, the smaller scale of nanobubbles may enable them to access the tumor extravascular compartment for imaging and therapy in closer proximity to cancer cells. Compelling preliminary demonstrations of the imaging and therapeutic abilities of nanobubbles have thus emerged, with emphasis on their ability to extravasate. However, studies to date rely on indirect histologic evidence that cannot confirm whether the structures remain intact beyond the vasculature - leaving their extravascular potential largely untapped. Methods: Nanobubble acoustic scattering was assessed using a recently reported ultra-stable formulation at low concentration (106 mL-1) and frequency (1 MHz), over a range of pressures (100-1500 kPa) in a channel phantom. The pressure-dependent response was utilized as a basis for in vivo experiments where ultrasound transmitters and receivers were integrated into a window chamber for simultaneous intravital multiphoton microscopy and acoustic monitoring in tumor-affected microcirculation. Microscopy and acoustic data were utilized to assess passive and active delivery of nanobubbles and determine whether they remained intact beyond the vasculature. Results: Nanobubbles exhibit pressure-dependent nonlinear acoustic scattering. Nanobubbles are also found to have prolonged acoustic vascular pharmacokinetics, and passively extravasate intact into tumors. Ultrasound stimulation of nanobubbles is shown to actively enhance the delivery of both intact nanobubbles and shell material, increasing their spatial bioavailability deeper into the extravascular space. A range of acute vascular effects were also observed. Conclusion: This study presents the first direct evidence that nanobubbles passively and actively extravasate intact in tumor tissue, and is the first to directly capture acute vascular events from ultrasound-stimulation of nanobubbles. The insights gained here demonstrate an important step towards unlocking the potential of nanobubbles and extending ultrasound-based applications.
Collapse
|
24
|
Abstract
The use of contrast agents as signal enhancers during ultrasound improves visualization and the diagnostic utility of this technology in medical imaging. Although widely used in many disciplines, contrast ultrasound is not routinely implemented in obstetrics, largely due to safety concerns of administered agents for pregnant women and the limited number of studies that address this issue. Here the microbubble characteristics that make them beneficial for enhancement of the blood pool and the quantification of real-time imaging are reviewed. Literature from pregnant animal model studies and safety assessments are detailed, and the potential for contrast-enhanced ultrasound to provide clinically relevant data and benefit our understanding of early placental development and detection of placental dysfunction is discussed.
Collapse
|
25
|
Crist RM, Dasa SSK, Liu CH, Clogston JD, Dobrovolskaia MA, Stern ST. Challenges in the development of nanoparticle-based imaging agents: Characterization and biology. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1665. [PMID: 32830448 DOI: 10.1002/wnan.1665] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022]
Abstract
Despite imaging agents being some of the earliest nanomedicines in clinical use, the vast majority of current research and translational activities in the nanomedicine field involves therapeutics, while imaging agents are severely underrepresented. The reasons for this lack of representation are several fold, including difficulties in synthesis and scale-up, biocompatibility issues, lack of suitable tissue/disease selective targeting ligands and receptors, and a high bar for regulatory approval. The recent focus on immunotherapies and personalized medicine, and development of nanoparticle constructs with better tissue distribution and selectivity, provide new opportunities for nanomedicine imaging agent development. This manuscript will provide an overview of trends in imaging nanomedicine characterization and biocompatibility, and new horizons for future development. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Rachael M Crist
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Siva Sai Krishna Dasa
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Christina H Liu
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland, USA
| | - Jeffrey D Clogston
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Stephan T Stern
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland, USA
| |
Collapse
|
26
|
Kehinde BA, Sharma P, Kaur S. Recent nano-, micro- and macrotechnological applications of ultrasonication in food-based systems. Crit Rev Food Sci Nutr 2020; 61:599-621. [PMID: 32208850 DOI: 10.1080/10408398.2020.1740646] [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] [Indexed: 01/02/2023]
Abstract
There is a neoteric and rising demand for nutritional and functional foods which behooves food processors to adopt processing techniques with optimal conservation of bioactive components in foods and with minimal pernicious impacts on the environment. Ultrasonication, a mechanochemical technique has proven to be an efficacious panacea to these concerns. In this review, an analytic exploration of recent researches and designs regarding ultrasound methodology and equipment on diverse food systems, technological scales, procedural parameters and outcomes of such experimentations optimally scrutinized. The relative effects of ultrasonication on food formulations, components and attributes such as nanoemulsions, nanocapsules, proteins, micronutrients, sensory and mechanical characteristics are evaluatively delineated. In food systems where ultrasonication was employed, it was found to have a remarkable effect on one or more quality parameters. This review is a supplementation to the pedagogical awareness to scholars on the suitability of ultrasonication for research procedures, and a call to industrial food brands on the adoption of this technique for the development of foods with optimally sustained nutrient profiles.
Collapse
Affiliation(s)
- Bababode Adesegun Kehinde
- Department of Biosystems and Agricultural Engineering, University of Kentucky, Lexington, Kentucky, USA
| | - Poorva Sharma
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, Punjab, India
| | - Sawinder Kaur
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
27
|
Nio AQX, Faraci A, Christensen-Jeffries K, Raymond JL, Monaghan MJ, Fuster D, Forsberg F, Eckersley RJ, Lamata P. Optimal Control of SonoVue Microbubbles to Estimate Hydrostatic Pressure. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2020; 67:557-567. [PMID: 31634833 PMCID: PMC7053253 DOI: 10.1109/tuffc.2019.2948759] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/16/2019] [Indexed: 05/13/2023]
Abstract
The measurement of cardiac and aortic pressures enables diagnostic insight into cardiac contractility and stiffness. However, these pressures are currently assessed invasively using pressure catheters. It may be possible to estimate these pressures less invasively by applying microbubble ultrasound contrast agents as pressure sensors. The aim of this study was to investigate the subharmonic response of the microbubble ultrasound contrast agent SonoVue (Bracco Spa, Milan, Italy) at physiological pressures using a static pressure phantom. A commercially available cell culture cassette with Luer connections was used as a static pressure chamber. SonoVue was added to the phantom, and radio frequency data were recorded on the ULtrasound Advanced Open Platform (ULA-OP). The mean subharmonic amplitude over a 40% bandwidth was extracted at 0-200-mmHg hydrostatic pressures, across 1.7-7.0-MHz transmit frequencies and 3.5%-100% maximum scanner acoustic output. The Rayleigh-Plesset equation for single-bubble oscillations and additional hysteresis experiments were used to provide insight into the mechanisms underlying the subharmonic pressure response of SonoVue. The subharmonic amplitude of SonoVue increased with hydrostatic pressure up to 50 mmHg across all transmit frequencies and decreased thereafter. A decreasing microbubble surface tension may drive the initial increase in the subharmonic amplitude of SonoVue with hydrostatic pressure, while shell buckling and microbubble destruction may contribute to the subsequent decrease above 125-mmHg pressure. In conclusion, a practical operating regime that may be applied to estimate cardiac and aortic blood pressures from the subharmonic signal of SonoVue has been identified.
Collapse
Affiliation(s)
- Amanda Q. X. Nio
- Department of Biomedical EngineeringSchool of Biomedical Engineering and Imaging SciencesKing’s College LondonLondonSE1 7EHU.K.
| | - Alessandro Faraci
- Department of Biomedical EngineeringSchool of Biomedical Engineering and Imaging SciencesKing’s College LondonLondonSE1 7EHU.K.
| | - Kirsten Christensen-Jeffries
- Department of Biomedical EngineeringSchool of Biomedical Engineering and Imaging SciencesKing’s College LondonLondonSE1 7EHU.K.
| | - Jason L. Raymond
- Department of Engineering ScienceUniversity of OxfordOxfordOX1 3PJU.K.
| | - Mark J. Monaghan
- Department of CardiologyKing’s College HospitalLondonSE5 9RSU.K.
| | - Daniel Fuster
- Institut Jean Le Rond D’Alembert, Sorbonne UniversitéCenter National de la Recherche Scientifique, UMR 7190F-75005ParisFrance
| | - Flemming Forsberg
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPA19107USA
| | - Robert J. Eckersley
- Department of Biomedical EngineeringSchool of Biomedical Engineering and Imaging SciencesKing’s College LondonLondonSE1 7EHU.K.
| | - Pablo Lamata
- Department of Biomedical EngineeringSchool of Biomedical Engineering and Imaging SciencesKing’s College LondonLondonSE1 7EHU.K.
| |
Collapse
|
28
|
Mantziaras G, Luvoni GC. Advanced ultrasound techniques in small animal reproduction imaging. Reprod Domest Anim 2020; 55 Suppl 2:17-25. [PMID: 31913545 DOI: 10.1111/rda.13587] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Abstract
Ultrasonography is the imaging technology of choice for the evaluation of the reproduction system and of pregnancy in both humans and animals. Over the past 10 years, there have been significant technological improvements of the equipment, while new technologies have been developed. Doppler, contrast-enhanced ultrasonography, elastography, and 3D/4D ultrasonography are advanced ultrasound techniques that have been designed as methods to increase the diagnostic sensitivity of two-dimensional (b-mode) ultrasound, and not as stand-alone tests. The basic physics as well as the advantages and limitations of these advanced ultrasound methods are briefly described. In the reproductive diagnostics of small animals, these techniques have gained an increased popularity as proved by the increased publication of several reports that are also briefly summarized in this review. Clinical applicability is to date limited because of a lack of research on the diagnostic value in concrete situations. Future research projects should focus also on standardization of the used techniques, on determination of thresholds to discriminate between healthy or diseases or fertile versus infertile and on the predictive value of advanced ultrasound findings. Continuing development and optimization of different ultrasound techniques as well as the increase of related scientific interest and worldwide research promises that the clinical interest on the use of advanced ultrasound techniques will increase in future.
Collapse
Affiliation(s)
| | - Gaia Cecilia Luvoni
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
29
|
Kee ALY, Teo BM. Biomedical applications of acoustically responsive phase shift nanodroplets: Current status and future directions. ULTRASONICS SONOCHEMISTRY 2019; 56:37-45. [PMID: 31101274 DOI: 10.1016/j.ultsonch.2019.03.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/08/2019] [Accepted: 03/22/2019] [Indexed: 05/19/2023]
Abstract
The evolution of ultrasonic contrast agents to enhance the reflectivity of structures in the human body has consolidated ultrasound's stance as a reliable diagnostic imaging modality. A significant development within this field includes the advent of liquid nanodroplets that are capable of vaporising into gaseous microbubbles upon ultrasonic irradiation. This literature review will therefore appraise and summarise the available literature on the generation of phase-shift nanodroplets, their formulations, applications, safety issues, future developments and any implications that may inhibit their clinical implementation. The main findings of this review affirm that phase change nanodroplets do indeed demonstrate functionality in drug delivery and targeting and characterisation of tumours. Its bioeffects however, have not yet been extensively researched, prompting further exploration into how bubble size can be controlled once it has vaporised into microbubbles and the resulting complications. As such, future research should be directed towards determining the safety, longevity and suitability of phase-shift nanodroplets over contrast agents in current clinical use.
Collapse
Affiliation(s)
- Allison Loo Yong Kee
- Department of Medical Imaging and Radiation Sciences, Monash University, 10 Chancellors Walk, Clayton, Victoria 3800, Australia
| | - Boon Mian Teo
- School of Chemistry, Monash University, 19 Rainforest Walk, Clayton, Victoria 3800, Australia.
| |
Collapse
|
30
|
Kanoulas E, Butler M, Rowley C, Voulgaridou V, Diamantis K, Duncan WC, McNeilly A, Averkiou M, Wijkstra H, Mischi M, Wilson RS, Lu W, Sboros V. Super-Resolution Contrast-Enhanced Ultrasound Methodology for the Identification of In Vivo Vascular Dynamics in 2D. Invest Radiol 2019; 54:500-516. [PMID: 31058661 PMCID: PMC6661242 DOI: 10.1097/rli.0000000000000565] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The aim of this study was to provide an ultrasound-based super-resolution methodology that can be implemented using clinical 2-dimensional ultrasound equipment and standard contrast-enhanced ultrasound modes. In addition, the aim is to achieve this for true-to-life patient imaging conditions, including realistic examination times of a few minutes and adequate image penetration depths that can be used to scan entire organs without sacrificing current super-resolution ultrasound imaging performance. METHODS Standard contrast-enhanced ultrasound was used along with bolus or infusion injections of SonoVue (Bracco, Geneva, Switzerland) microbubble (MB) suspensions. An image analysis methodology, translated from light microscopy algorithms, was developed for use with ultrasound contrast imaging video data. New features that are tailored for ultrasound contrast image data were developed for MB detection and segmentation, so that the algorithm can deal with single and overlapping MBs. The method was tested initially on synthetic data, then with a simple microvessel phantom, and then with in vivo ultrasound contrast video loops from sheep ovaries. Tracks detailing the vascular structure and corresponding velocity map of the sheep ovary were reconstructed. Images acquired from light microscopy, optical projection tomography, and optical coherence tomography were compared with the vasculature network that was revealed in the ultrasound contrast data. The final method was applied to clinical prostate data as a proof of principle. RESULTS Features of the ovary identified in optical modalities mentioned previously were also identified in the ultrasound super-resolution density maps. Follicular areas, follicle wall, vessel diameter, and tissue dimensions were very similar. An approximately 8.5-fold resolution gain was demonstrated in vessel width, as vessels of width down to 60 μm were detected and verified (λ = 514 μm). Best agreement was found between ultrasound measurements and optical coherence tomography with 10% difference in the measured vessel widths, whereas ex vivo microscopy measurements were significantly lower by 43% on average. The results were mostly achieved using video loops of under 2-minute duration that included respiratory motion. A feasibility study on a human prostate showed good agreement between density and velocity ultrasound maps with the histological evaluation of the location of a tumor. CONCLUSIONS The feasibility of a 2-dimensional contrast-enhanced ultrasound-based super-resolution method was demonstrated using in vitro, synthetic and in vivo animal data. The method reduces the examination times to a few minutes using state-of-the-art ultrasound equipment and can provide super-resolution maps for an entire prostate with similar resolution to that achieved in other studies.
Collapse
Affiliation(s)
- Evangelos Kanoulas
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| | - Mairead Butler
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| | - Caitlin Rowley
- Department of Physics, Heriot-Watt University, Riccarton
| | - Vasiliki Voulgaridou
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| | | | - William Colin Duncan
- Center for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan McNeilly
- Center for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Massimo Mischi
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; and
| | - Rhodri Simon Wilson
- **Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Weiping Lu
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| | - Vassilis Sboros
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| |
Collapse
|
31
|
Velasquez M, Emerson MG, Diaz E, Kennedy W, Rubesova E, Barth RA. The learning curve of contrast-enhanced 'microbubble' voiding urosonography-validation study. J Pediatr Urol 2019; 15:385.e1-385.e6. [PMID: 31133505 DOI: 10.1016/j.jpurol.2019.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/15/2019] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Vesicoureteral reflux (VUR) is a common pediatric urologic condition associated with urinary tract infection and pyelonephritis. It can be diagnosed via fluoroscopic voiding cystourethrogram (VCUG) and, more recently, contrast-enhanced voiding ultrasonography (ceVUS), which does not expose the patient to ionizing radiation. Voiding urosonography contrast agents used for the diagnosis of VUR have been widely available in Europe but were approved by the Food and Drug Administration for use in the United States only in 2016. OBJECTIVE The objective was to optimize a protocol and compare the diagnostic performance of ceVUS to fluoroscopic VCUG in an academic medical center naïve to previous use of contrast-enhanced voiding urosonography. STUDY DESIGN Thirty-nine patients referred for clinically indicated evaluation of VUR were enrolled between September 2016 and March 2017. Patients underwent contrast-enhanced ultrasonography with prediluted Lumason and under the same catheterization underwent fluoroscopic VCUG. Comparative grading was performed by pediatric radiologists on-site at the time of examination. RESULTS Reflux was observed in 16 of 39 patients (20 of 64 renal units) ranging from grades 1 through 5. VCUG and ceVUS were concordant for detecting reflux in 10 of 39 patients (14 of 84 renal units) and excluding reflux in 23 of 39 patients (64 of 84 renal units) (Fig. 1). Using contrast enhanced voiding urosonography, 1 of 20 renal units had high-grade and 2 of 20 renal units had low-grade reflux that was not found on fluoroscopy. Using fluoroscopy, 1 of 20 renal units had high-grade and 2 of 20 renal units had low-grade reflux that had not been found on ceVUS. Two of 20 renal units were upgraded from low-grade on ceVUS to high-grade on fluoroscopy. This corresponds to a Cohen's kappa of 0.72 (confidence interval [CI] 0.54-0.91) or 'moderate.' DISCUSSION During our investigation, we noted that there was a technical learning curve related to poor contrast mixing and the need to titrate the concentration of Lumason. However, over the course of the study, we were able to correct the technical aspects. Ultimately, our results showed good correlation between VCUG and Lumason ceVUS and only slightly less correlation than published studies by experienced centers. Future studies with voiding should allow for improved urethral visualization. CONCLUSION While there is a considerable learning curve to the implementation of ceVUS for the diagnosis of pediatric VUR, these technical aspects can be corrected. Even a center previously naïve to contrast-enhanced ultrasound technology can, over a short period of time, demonstrate good correlation between VCUG and ceVUS in the diagnosis of VUR. Translation of ceVUS into clinical practice is an alternative to VCUG for diagnosis of reflux, is feasible, and can eliminate the radiation exposure associated with a VCUG.
Collapse
Affiliation(s)
- M Velasquez
- Stanford Children's Health (Lucile Packard Children's Hospital), Department of Urology, Stanford University School of Medicine, 725 Welch Road - Room 1890, Stanford, CA 94304-5913, USA
| | - M G Emerson
- Radiology - Diagnostic, 677 N Wilmot Rd, Tucson, AZ 85711, USA
| | - E Diaz
- Stanford Children's Health (Lucile Packard Children's Hospital), Department of Urology, Stanford University School of Medicine, 725 Welch Road - Room 1890, Stanford, CA 94304-5913, USA
| | - W Kennedy
- Stanford Children's Health (Lucile Packard Children's Hospital), Department of Urology, Stanford University School of Medicine, 725 Welch Road - Room 1890, Stanford, CA 94304-5913, USA
| | - E Rubesova
- Stanford Children's Health (Lucile Packard Children's Hospital), Department of Radiology, Stanford University School of Medicine, 725 Welch Road - Room 1890, Stanford, CA 94304-5913, USA
| | - R A Barth
- Stanford Children's Health (Lucile Packard Children's Hospital), Department of Radiology, Stanford University School of Medicine, 725 Welch Road - Room 1890, Stanford, CA 94304-5913, USA.
| |
Collapse
|
32
|
Revels JW, Dey CB, Aggarwal A, London SS, Katz D, Menias C, Moshiri M. More Than Just 2 Layers: A Comprehensive Multimodality Imaging Review of Endometrial Abnormalities. Curr Probl Diagn Radiol 2019; 49:431-446. [PMID: 31307863 DOI: 10.1067/j.cpradiol.2019.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/06/2019] [Accepted: 06/25/2019] [Indexed: 11/22/2022]
Abstract
Endometrial abnormalities develop in female patients of all ages. Symptoms related to endometrial pathologies are among the most common causes of gynecologist office visits, with the radiologists playing an important role in endometrial evaluation. In some instances, the radiologist may be the first physician to note endometrial pathology. In this article, we will provide a comprehensive review of radiologic modalities utilized in the evaluation of the endometrium, as well as the imaging appearance of various endometrial disease processes.
Collapse
Affiliation(s)
| | - Courtney B Dey
- Department of Radiology, Eastern Virginia Medical School, Sentara Norfolk General Hospital, Norfolk, VA
| | - Abhi Aggarwal
- Department of Radiology, Eastern Virginia Medical School, Sentara Norfolk General Hospital, Norfolk, VA
| | - Sean S London
- Department of Radiology, University of Washington, Seattle, WA
| | - Douglas Katz
- Department of Radiology, NYU Winthrop Hospital, Mineola, NY
| | | | - Mariam Moshiri
- Department of Radiology, University of Washington, Seattle, WA
| |
Collapse
|
33
|
Maruf A, Wang Y, Yin T, Huang J, Wang N, Durkan C, Tan Y, Wu W, Wang G. Atherosclerosis Treatment with Stimuli-Responsive Nanoagents: Recent Advances and Future Perspectives. Adv Healthc Mater 2019; 8:e1900036. [PMID: 30945462 DOI: 10.1002/adhm.201900036] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/06/2019] [Indexed: 01/04/2023]
Abstract
Atherosclerosis is the root of approximately one-third of global mortalities. Nanotechnology exhibits splendid prospects to combat atherosclerosis at the molecular level by engineering smart nanoagents with versatile functionalizations. Significant advances in nanoengineering enable nanoagents to autonomously navigate in the bloodstream, escape from biological barriers, and assemble with their nanocohort at the targeted lesion. The assembly of nanoagents with endogenous and exogenous stimuli breaks down their shells, facilitates intracellular delivery, releases their cargo to kill the corrupt cells, and gives imaging reports. All these improvements pave the way toward personalized medicine for atherosclerosis. This review systematically summarizes the recent advances in stimuli-responsive nanoagents for atherosclerosis management and its progress in clinical trials.
Collapse
Affiliation(s)
- Ali Maruf
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Tieyin Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Junli Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Nan Wang
- The Nanoscience CentreUniversity of Cambridge Cambridge CB3 0FF UK
| | - Colm Durkan
- The Nanoscience CentreUniversity of Cambridge Cambridge CB3 0FF UK
| | - Youhua Tan
- Department of Biomedical EngineeringThe Hong Kong Polytechnic University Hong Kong SAR 999077 China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| |
Collapse
|
34
|
Lång K, Arboleda C, Forte S, Wang Z, Prevrhal S, Koehler T, Kuhn N, David B, Jefimovs K, Kubik-Huch RA, Stampanoni M. Microbubbles as a contrast agent in grating interferometry mammography: an ex vivo proof-of-mechanism study. Eur Radiol Exp 2019; 3:19. [PMID: 31115796 PMCID: PMC6529489 DOI: 10.1186/s41747-019-0097-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/04/2019] [Indexed: 11/24/2022] Open
Abstract
Grating interferometry mammography (GIM) is an experimental breast imaging method at the edge of being clinically implemented. Besides attenuation, GIM can measure the refraction and scattering of x-rays resulting in differential phase contrast (DPC) and dark-field (DF) images. In this exploratory study, we assessed the feasibility of using microbubbles as a contrast agent in GIM. Two millilitres of microbubbles and iodine were respectively injected into ex vivo breast phantoms, consisting of fresh chicken breasts. Native and postcontrast images were acquired with a clinically compatible GIM setup, operated at 38 kVp, 14-s acquisition time, and with a dose of 1.3 mGy. The visibility of the contrast agents was analysed in a side-by-side comparison by three radiologists. The contrast-to-noise-ratio (CNR) was calculated for each contrast agent. We found that both contrast agents were judged to be visible by the readers. The mean CNR was 3.1 ± 1.9 for microbubbles in DF and 24.2 ± 6.5 for iodine in attenuation. In conclusion, this is a first proof-of-mechanism study that microbubbles could be used as a contrast agent in clinically compatible GIM, due to their scattering properties, which implies the potential use of a contrast agent with a high safety profile in x-ray-based breast imaging.
Collapse
Affiliation(s)
- Kristina Lång
- Swiss Light Source, ETH Zurich, Paul Scherrer Institute, 5232, Villigen, Switzerland. .,Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland.
| | - Carolina Arboleda
- Swiss Light Source, ETH Zurich, Paul Scherrer Institute, 5232, Villigen, Switzerland.,Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Serafino Forte
- Department of Radiology, Kantonsspital Baden, Im Ergel 1, 5404, Baden, Switzerland
| | - Zhentian Wang
- Swiss Light Source, ETH Zurich, Paul Scherrer Institute, 5232, Villigen, Switzerland.,Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Sven Prevrhal
- Philips GmbH Innovative Technologies, Research Laboratories, Philips Research Hamburg, Röntgenstrasse 24-26, 22335, Hamburg, Germany
| | - Thomas Koehler
- Philips GmbH Innovative Technologies, Research Laboratories, Philips Research Hamburg, Röntgenstrasse 24-26, 22335, Hamburg, Germany
| | - Norbert Kuhn
- Philips GmbH Innovative Technologies, Research Laboratories, Philips Research Hamburg, Röntgenstrasse 24-26, 22335, Hamburg, Germany
| | - Bernd David
- Philips GmbH Innovative Technologies, Research Laboratories, Philips Research Hamburg, Röntgenstrasse 24-26, 22335, Hamburg, Germany
| | - Konstantins Jefimovs
- Swiss Light Source, ETH Zurich, Paul Scherrer Institute, 5232, Villigen, Switzerland.,Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Rahel A Kubik-Huch
- Department of Radiology, Kantonsspital Baden, Im Ergel 1, 5404, Baden, Switzerland
| | - Marco Stampanoni
- Swiss Light Source, ETH Zurich, Paul Scherrer Institute, 5232, Villigen, Switzerland.,Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Koruk H, Choi JJ. Displacement of a bubble located at a fluid-viscoelastic medium interface. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2019; 145:EL410. [PMID: 31153355 DOI: 10.1121/1.5108678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/29/2019] [Indexed: 06/09/2023]
Abstract
A model for estimating the displacement of a bubble located at a fluid-viscoelastic medium interface in response to acoustic radiation force is presented by extending the model for a spherical object embedded in a bulk material. The effects of the stiffness and viscosity of the viscoelastic medium and the amplitude and duration of the excitation force on bubble displacement were investigated using the proposed model. The results show that bubble displacement has a nonlinear relationship with excitation duration and viscosity. The time at which the steady state is reached increases with increasing medium viscosity and decreasing medium stiffness.
Collapse
Affiliation(s)
- Hasan Koruk
- Mechanical Engineering Department, MEF University, Istanbul 34396,
| | - James J Choi
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United
| |
Collapse
|
36
|
Kumar S, Purtell C, Peterson A, Gibbons P, Khan AM, Heitner SB. Safety profile of ultrasound enhancing agents in echocardiography. Echocardiography 2019; 36:1041-1044. [PMID: 31038768 DOI: 10.1111/echo.14344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/12/2019] [Accepted: 03/30/2019] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Ultrasound enhancing agents (UEAs) are often utilized to enhance ultrasound image quality; however, concerns about adverse reactions have limited their use. Moreover, these agents had been either contraindicated or are labeled with a warning in patients with intra-cardiac shunts because of a theoretic risk of systemic microvascular obstruction. This labeling was recently removed in the United States, but data in these patients are lacking. METHODS Over a 15-month period, patients receiving three different FDA-approved UEAs at our center were prospectively evaluated for clinically significant adverse events (AEs). RESULTS A total of 5521 UEA administrations were performed (Definity® : 3306, Lumason® : 2137, Optison® : 78). There were 14 AEs (0.25%) reported (Lumason® : 0.05% [n = 1] vs Definity® : 0.39% [n = 13], P = 0.02). Back pain was the most common complaint (n = 9), followed by headache (n = 2), rash (n = 2), dyspnea (n = 2), and palpitations (n = 1). Among the 33 patients known to have intra-cardiac shunts, there were no AEs. Known right-to-left shunts with positive saline bubble study were present in 20 patients (Lumason® : n = 9, Definity® : n = 11). Left-to-right atrial shunts based on color Doppler were present in 10 patients (Lumason® : n = 5, Definity® n = 5). Three patients were known to have ventricular septal defect with left-to-right flow (Definity® : n = 2, Optison® : n = 1). CONCLUSION Adverse events were significantly higher with Definity® ; however, overall incidences were low, and AEs were minor. Furthermore, no AEs were reported in patients with known intra-cardiac shunts. UEAs showed a good safety profile in our study and should be afforded to all appropriate patients, including those with known intra-cardiac shunts.
Collapse
Affiliation(s)
- Suwen Kumar
- Oregon Health and Science University, Portland, Oregon
| | - Chris Purtell
- Oregon Health and Science University, Portland, Oregon
| | | | - Paul Gibbons
- Oregon Health and Science University, Portland, Oregon
| | | | | |
Collapse
|
37
|
Beguin E, Bau L, Shrivastava S, Stride E. Comparing Strategies for Magnetic Functionalization of Microbubbles. ACS APPLIED MATERIALS & INTERFACES 2019; 11:1829-1840. [PMID: 30574777 DOI: 10.1021/acsami.8b18418] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The advancement of ultrasound-mediated therapy has stimulated the development of drug-loaded microbubble agents that can be targeted to a region of interest through an applied magnetic field prior to ultrasound activation. However, the need to incorporate therapeutic molecules while optimizing the responsiveness to both magnetic and acoustic fields and maintaining adequate stability poses a considerable challenge for microbubble synthesis. The aim of this study was to evaluate three different methods for incorporating iron oxide nanoparticles (IONPs) into phospholipid-coated microbubbles using (1) hydrophobic IONPs within an oil layer below the microbubble shell, (2) phospholipid-stabilized IONPs within the shell, or (3) hydrophilic IONPs noncovalently bound to the surface of the microbubble. All microbubbles exhibited similar acoustic response at both 1 and 7 MHz. The half-life of the microbubbles was more than doubled by the addition of IONPs by using both surface and phospholipid-mediated loading methods, provided the lipid used to coat the IONPs was the same as that constituting the microbubble shell. The highest loading of IONPs per microbubble was also achieved with the surface loading method, and these microbubbles were the most responsive to an applied magnetic field, showing a 3-fold increase in the number of retained microbubbles compared to other groups. For the purpose of drug delivery, surface loading of IONPs could restrict the attachment of hydrophilic drugs to the microbubble shell, but hydrophobic drugs could still be incorporated. In contrast, although the incorporation of phospholipid IONPs produced more weakly magnetic microbubbles, it would not interfere with hydrophilic drug loading on the surface of the microbubble.
Collapse
Affiliation(s)
- Estelle Beguin
- Department of Engineering Science, Institute of Biomedical Engineering , University of Oxford , Oxford OX3 7DQ , U.K
| | - Luca Bau
- Department of Engineering Science, Institute of Biomedical Engineering , University of Oxford , Oxford OX3 7DQ , U.K
| | - Shamit Shrivastava
- Department of Engineering Science, Institute of Biomedical Engineering , University of Oxford , Oxford OX3 7DQ , U.K
| | - Eleanor Stride
- Department of Engineering Science, Institute of Biomedical Engineering , University of Oxford , Oxford OX3 7DQ , U.K
| |
Collapse
|
38
|
Patnaik SS, Simionescu DT, Goergen CJ, Hoyt K, Sirsi S, Finol EA. Pentagalloyl Glucose and Its Functional Role in Vascular Health: Biomechanics and Drug-Delivery Characteristics. Ann Biomed Eng 2019; 47:39-59. [PMID: 30298373 PMCID: PMC6318003 DOI: 10.1007/s10439-018-02145-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/28/2018] [Indexed: 02/08/2023]
Abstract
Pentagalloyl glucose (PGG) is an elastin-stabilizing polyphenolic compound that has significant biomedical benefits, such as being a free radical sink, an anti-inflammatory agent, anti-diabetic agent, enzymatic resistant properties, etc. This review article focuses on the important benefits of PGG on vascular health, including its role in tissue mechanics, the different modes of pharmacological administration (e.g., oral, intravenous and endovascular route, intraperitoneal route, subcutaneous route, and nanoparticle based delivery and microbubble-based delivery), and its potential therapeutic role in vascular diseases such as abdominal aortic aneurysms (AAA). In particular, the use of PGG for AAA suppression and prevention has been demonstrated to be effective only in the calcium chloride rat AAA model. Therefore, in this critical review we address the challenges that lie ahead for the clinical translation of PGG as an AAA growth suppressor.
Collapse
Affiliation(s)
- Sourav S Patnaik
- Vascular Biomechanics and Biofluids Laboratory, Department of Mechanical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249-0670, USA
| | - Dan T Simionescu
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shashank Sirsi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ender A Finol
- Vascular Biomechanics and Biofluids Laboratory, Department of Mechanical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249-0670, USA.
| |
Collapse
|
39
|
Kauscher U, Holme MN, Björnmalm M, Stevens MM. Physical stimuli-responsive vesicles in drug delivery: Beyond liposomes and polymersomes. Adv Drug Deliv Rev 2019; 138:259-275. [PMID: 30947810 PMCID: PMC7180078 DOI: 10.1016/j.addr.2018.10.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/30/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022]
Abstract
Over the past few decades, a range of vesicle-based drug delivery systems have entered clinical practice and several others are in various stages of clinical translation. While most of these vesicle constructs are lipid-based (liposomes), or polymer-based (polymersomes), recently new classes of vesicles have emerged that defy easy classification. Examples include assemblies with small molecule amphiphiles, biologically derived membranes, hybrid vesicles with two or more classes of amphiphiles, or more complex hierarchical structures such as vesicles incorporating gas bubbles or nanoparticulates in the lumen or membrane. In this review, we explore these recent advances and emerging trends at the edge and just beyond the research fields of conventional liposomes and polymersomes. A focus of this review is the distinct behaviors observed for these classes of vesicles when exposed to physical stimuli - such as ultrasound, heat, light and mechanical triggers - and we discuss the resulting potential for new types of drug delivery, with a special emphasis on current challenges and opportunities.
Collapse
Affiliation(s)
- Ulrike Kauscher
- Department of Materials, Imperial College London, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Margaret N Holme
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Mattias Björnmalm
- Department of Materials, Imperial College London, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Molly M Stevens
- Department of Materials, Imperial College London, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-171 77, Sweden.
| |
Collapse
|
40
|
Alrashed A, Ahmad H, Khalili K, Kim TK, Jang HJ, Atri M. Negative Predictive Value of Contrast-Enhanced Ultrasound in Differentiating Avascular Solid-Appearing From Vascularized Masses: A Retrospective Consecutive Study. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2018; 37:2935-2942. [PMID: 29761521 DOI: 10.1002/jum.14659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVES To determine the negative predictive value (NPV) of contrast-enhanced ultrasound (CEUS) to establish the lack of vascularity in a mass. METHODS This work was an Institutional Review Board-approved, Health Insurance Portability and Accountability Act-compliant retrospective study. Acquisition of consent was waived. We included all CEUS examinations performed for tissue characterization between 2004 and 2014 that reported showing no vascularity in a mass. Contrast-enhanced ultrasound findings were considered true-negative when there was stability on imaging for at least 1 year or no evidence of a solid mass, if biopsied, and false-negative if there was lesion growth on imaging within 12 months or an indication of a solid mass on the pathologic examination, if biopsied. One author reviewed all of the reports and follow-up examinations. We conducted a consensus review of all false-negative findings mixed with an equal number of true-negative findings by 2 reviewers, who were blinded to the final results. RESULTS The study population consisted of 97 CEUS examinations in 97 patients, including 48 women and 49 men (mean age ± SD, 65 ± 14 years). Examinations were performed for lesion characterization in the liver (n = 23), pancreas (n = 17), kidney (n = 54), 1 gallbladder, 1 adnexa, and 1 peritoneal lesion. The overall false-negative rate on the official prospective review was 2% (2 of 97). Two false-negative findings were correctly identified on the consensus review. The NPV of CEUS was 97.9% (95 of 97; 95% confidence interval, 93%- 99%) on the official review. CONCLUSIONS Contrast-enhanced ultrasound has a very high NPV to exclude the presence of flow in a mass, and it can be used to exclude the presence of a solid mass.
Collapse
Affiliation(s)
| | - Hassan Ahmad
- Joint Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Korosh Khalili
- Joint Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Tae Kyoung Kim
- Joint Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Huynh-Jung Jang
- Joint Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Mostafa Atri
- Joint Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Borden MA, Song KH. Reverse engineering the ultrasound contrast agent. Adv Colloid Interface Sci 2018; 262:39-49. [PMID: 30396507 PMCID: PMC6268001 DOI: 10.1016/j.cis.2018.10.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/09/2018] [Accepted: 10/22/2018] [Indexed: 12/31/2022]
Abstract
In this review, a brief history and current state-of-the-art is given to stimulate the rational design of new microbubbles through the reverse engineering of current ultrasound contrast agents (UCAs). It is shown that an effective microbubble should be biocompatible, echogenic and stable. Physical mechanisms and engineering calculations have been provided to illustrate these properties and how they can be achieved. The reverse-engineering design paradigm is applied to study current FDA-approved and commercially available UCAs. Given the sophistication of microbubble designs reported in the literature, rapid development and adoption of ultrasound device hardware and techniques, and the growing number of revolutionary biomedical applications moving toward the clinic, the field of Microbubble Engineering is fertile for breakthroughs in next-generation UCA technology. It is up to current and future microbubble engineers and clinicians to push forward with regulatory approval and clinical adoption of advanced UCA technologies in the years to come.
Collapse
Affiliation(s)
- Mark A Borden
- Mechanical Engineering, University of Colorado, Boulder, CO 80309-0427, USA.
| | - Kang-Ho Song
- Mechanical Engineering, University of Colorado, Boulder, CO 80309-0427, USA
| |
Collapse
|
42
|
Blackmore DG, Turpin F, Mohamed AZ, Zong F, Pandit R, Pelekanos M, Nasrallah F, Sah P, Bartlett PF, Götz J. Multimodal analysis of aged wild-type mice exposed to repeated scanning ultrasound treatments demonstrates long-term safety. Am J Cancer Res 2018; 8:6233-6247. [PMID: 30613294 PMCID: PMC6299703 DOI: 10.7150/thno.27941] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier presents a major challenge for the delivery of therapeutic agents to the brain; however, it can be transiently opened by combining low intensity ultrasound with microbubble infusion. Studies evaluating this technology have largely been performed in rodents, including models of neurological conditions. However, despite promising outcomes in terms of drug delivery and the amelioration of neurological impairments, the potential for long-term adverse effects presents a major concern in the context of clinical applications. Methods: To fill this gap, we repeatedly treated 12-month-old wild-type mice with ultrasound, followed by a multimodal analysis for up to 18 months of age. Results: We found that spatial memory in these aged mice was not adversely affected as assessed in the active place avoidance test. Sholl analysis of Golgi impregnations in the dentate gyrus of the hippocampus did not reveal any changes to the neuronal cytoarchitecture. Long-term potentiation, a cellular correlate of memory, was still achievable, magnetic resonance spectroscopy revealed no major changes in metabolites, and diffusion tensor imaging revealed normal microstructure and tissue integrity in the hippocampus. More specifically, all measures of diffusion appeared to support a neuroprotective effect of ultrasound treatment on the brain. Conclusion: This multimodal analysis indicates that therapeutic ultrasound for blood-brain barrier opening is safe and potentially protective in the long-term, underscoring its validity as a potential treatment modality for diseases of the brain.
Collapse
|
43
|
Ashrafi AN, Nassiri N, Gill IS, Gulati M, Park D, de Castro Abreu AL. Contrast-Enhanced Transrectal Ultrasound in Focal Therapy for Prostate Cancer. Curr Urol Rep 2018; 19:87. [PMID: 30155585 PMCID: PMC9084632 DOI: 10.1007/s11934-018-0836-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Contrast-enhanced transrectal ultrasound (CeTRUS) is an emerging imaging technique in prostate cancer (PCa) diagnosis and treatment. We review the utility and implications of CeTRUS in PCa focal therapy (FT). RECENT FINDINGS CeTRUS utilizes intravenous injection of ultrasound-enhancing agents followed by high-resolution ultrasound to evaluate tissue microvasculature and differentiate between benign tissue and PCa, with the latter demonstrating increased enhancement. The potential utility of CeTRUS in FT for PCa extends to pre-, intra- and post-operative settings. CeTRUS may detect PCa, facilitate targeted biopsy and aid surgical planning prior to FT. During FT, the treated area can be visualized as a well-demarcated non-enhancing zone and continuous real-time assessment allows immediate re-treatment if necessary. Following FT, the changes on CeTRUS are immediate and consistent, thus facilitating repeat imaging for comparison during follow-up. Areas suspicious for recurrence may be detected and target-biopsied. Enhancement can be quantified using time-intensity curves allowing objective assessment and comparison. Based on encouraging early outcomes, CeTRUS may become an alternative imaging modality in prostate cancer FT. Further study with larger cohorts and longer follow-up are needed.
Collapse
Affiliation(s)
- Akbar N Ashrafi
- USC Institute of Urology, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, Suite 7416, Los Angeles, CA, 90089, USA
| | - Nima Nassiri
- USC Institute of Urology, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, Suite 7416, Los Angeles, CA, 90089, USA
| | - Inderbir S Gill
- USC Institute of Urology, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, Suite 7416, Los Angeles, CA, 90089, USA
| | - Mittul Gulati
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel Park
- USC Institute of Urology, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, Suite 7416, Los Angeles, CA, 90089, USA
| | - Andre L de Castro Abreu
- USC Institute of Urology, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, Suite 7416, Los Angeles, CA, 90089, USA.
| |
Collapse
|
44
|
Guo JC, Yang YJ, Guo M, Jiang XL. Ultrasound-guided intertumoral injection of contrast agents combined with human p53 gene for the treatment of breast cancer. Kaohsiung J Med Sci 2018; 34:438-446. [PMID: 30041761 DOI: 10.1016/j.kjms.2018.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/12/2018] [Accepted: 03/19/2018] [Indexed: 01/15/2023] Open
Abstract
The objective of this study was to investigate the effects of ultrasound-guided injection of ultrasound contrast agents (UCAs) and the p53 gene on the treatment of rats with breast cancer (BC). Assembly of the p53 expression vector as well as that of a rat model with BC consisted of 200 successfully modeled rats randomly divided into 5 groups: p53 gene introduction, p53 gene introduction + ultrasound irradiation, p53 gene introduction + UCAs, p53 gene introduction + UCA + ultrasound irradiation, and UCA + ultrasound irradiation groups. Expression of p53 was detected via quantitative real-time polymerase chain reaction (qRT-PCR), western blotting and immunohistochemical staining. In the p53 gene introduction + ultrasound irradiation group, we observed increased tumor volume with blood flow signals around and necrotic tumor tissues with an inhibition rate of 36.30%, as well as higher expression of p53 than that in the p53 gene introduction group and p53 gene introduction + UCA group. In the p53 gene introduction + UCA + ultrasound irradiation group, tumor volume increased slightly with reduced blood flow signals and massive degenerative necrosis of tumor cells was identified with inhibition rate of 62.62%, and expression of p53 was higher than that in the rest groups. Taken together, the key findings obtained from the present study elucidate that injection of p53 gene and UCA microbubbles guided by ultrasound could increase the expression of p53, thus inhibiting the tumor growth in rats with BC.
Collapse
Affiliation(s)
- Jun-Cheng Guo
- Graduate School of Central South University, Chang Sha, 410083, PR China
| | - Yi-Jun Yang
- Hospital Dean's Office, Haikou People's Hospital, Haikou, PR China
| | - Min Guo
- Hainan General Hospital, Hainan Provincial Academy of Clinical Medical Sciences, Haikou 570311, PR China.
| | - Xiang-Ling Jiang
- Hainan General Hospital, Hainan Provincial Academy of Clinical Medical Sciences, Haikou 570311, PR China
| |
Collapse
|
45
|
Abstract
Contrast-enhanced ultrasound imaging is a recently approved technique in the United States that uses a specific contrast agent, namely, microbubbles, consisting mainly of a gas core and a stabilized biological shell. These compounds allow for the visualization of small vascular beds and improve characterization of anatomic structures and lesions. They have a relatively safe profile and are primarily excreted through the lungs.
Collapse
Affiliation(s)
- David Hunt
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Javier Romero
- Department of Radiology, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
46
|
Fix SM, Nyankima AG, McSweeney MD, Tsuruta JK, Lai SK, Dayton PA. Accelerated Clearance of Ultrasound Contrast Agents Containing Polyethylene Glycol is Associated with the Generation of Anti-Polyethylene Glycol Antibodies. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:1266-1280. [PMID: 29602540 PMCID: PMC6171506 DOI: 10.1016/j.ultrasmedbio.2018.02.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 02/10/2018] [Accepted: 02/12/2018] [Indexed: 05/10/2023]
Abstract
Emerging evidence suggests that the immune system can recognize polyethylene glycol (PEG), leading to the accelerated blood clearance (ABC) of PEGylated particles. Our aim here was to study the generation of anti-PEG immunity and changes in PEGylated microbubble pharmacokinetics during repeated contrast-enhanced ultrasound imaging in rats. We administered homemade PEGylated microbubbles multiple times over a 28-d period and observed dramatically accelerated clearance (4.2 × reduction in half-life), which was associated with robust anti-PEG IgM and anti-PEG IgG antibody production. Dosing animals with free PEG as a competition agent before homemade PEGylated microbubble administration significantly prolonged microbubble circulation, suggesting that ABC was largely driven by circulating anti-PEG antibodies. Experiments with U.S. Food and Drug Administration-approved Definity microbubbles similarly resulted in ABC and the generation of anti-PEG antibodies. Experiments repeated with non-PEGylated Optison microbubbles revealed a slight shift in clearance, indicating that immunologic factors beyond anti-PEG immunity may play a role in ABC, especially of non-PEGylated agents.
Collapse
Affiliation(s)
- Samantha M Fix
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - A Gloria Nyankima
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Morgan D McSweeney
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - James K Tsuruta
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Samuel K Lai
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paul A Dayton
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA.
| |
Collapse
|
47
|
Durot I, Wilson SR, Willmann JK. Contrast-enhanced ultrasound of malignant liver lesions. Abdom Radiol (NY) 2018; 43:819-847. [PMID: 29094174 DOI: 10.1007/s00261-017-1360-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Contrast-enhanced ultrasound (CEUS) is a safe, relatively inexpensive, and widely available imaging technique using dedicated imaging ultrasound sequences and FDA-approved contrast microbubbles that allow detection and characterization of malignant focal liver lesions with high diagnostic accuracy. CEUS provides dynamic real-time imaging with high spatial and temporal capability, allowing for unique contributions to the already established protocols for diagnosing focal liver lesions using CT and MR imaging. In patients with lesions indeterminate on CT and MRI, CEUS is a helpful problem-solving complementary tool that improves patient management. Furthermore, CEUS assists guidance of liver biopsies and local treatment. Variations of CEUS such as DCE-US and ultrasound molecular imaging are emerging for quantitative monitoring of treatment effects and possible earlier detection of cancer. In this review, basic principles of CEUS techniques and ultrasound contrast agents along with a description of the enhancement patterns of malignant liver lesions are summarized. Also, a discussion of the role of CEUS for treatment guidance and monitoring, intraoperative CEUS, and an outlook on emerging applications is provided.
Collapse
|
48
|
Dave JK, Mc Donald ME, Mehrotra P, Kohut AR, Eisenbrey JR, Forsberg F. Recent technological advancements in cardiac ultrasound imaging. ULTRASONICS 2018; 84:329-340. [PMID: 29223692 PMCID: PMC5808891 DOI: 10.1016/j.ultras.2017.11.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 10/27/2017] [Accepted: 11/20/2017] [Indexed: 05/07/2023]
Abstract
About 92.1 million Americans suffer from at least one type of cardiovascular disease. Worldwide, cardiovascular diseases are the number one cause of death (about 31% of all global deaths). Recent technological advancements in cardiac ultrasound imaging are expected to aid in the clinical diagnosis of many cardiovascular diseases. This article provides an overview of such recent technological advancements, specifically focusing on tissue Doppler imaging, strain imaging, contrast echocardiography, 3D echocardiography, point-of-care echocardiography, 3D volumetric flow assessments, and elastography. With these advancements ultrasound imaging is rapidly changing the domain of cardiac imaging. The advantages offered by ultrasound imaging include real-time imaging, imaging at patient bed-side, cost-effectiveness and ionizing-radiation-free imaging. Along with these advantages, the steps taken towards standardization of ultrasound based quantitative markers, reviewed here, will play a major role in addressing the healthcare burden associated with cardiovascular diseases.
Collapse
Affiliation(s)
- Jaydev K Dave
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Maureen E Mc Donald
- Department of Radiologic Sciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Praveen Mehrotra
- Division of Cardiology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew R Kohut
- Division of Cardiology, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
49
|
Porter TR, Mulvagh SL, Abdelmoneim SS, Becher H, Belcik JT, Bierig M, Choy J, Gaibazzi N, Gillam LD, Janardhanan R, Kutty S, Leong-Poi H, Lindner JR, Main ML, Mathias W, Park MM, Senior R, Villanueva F. Clinical Applications of Ultrasonic Enhancing Agents in Echocardiography: 2018 American Society of Echocardiography Guidelines Update. J Am Soc Echocardiogr 2018; 31:241-274. [DOI: 10.1016/j.echo.2017.11.013] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
50
|
Peng Y, Seekell RP, Cole AR, Lamothe JR, Lock AT, van den Bosch S, Tang X, Kheir JN, Polizzotti BD. Interfacial Nanoprecipitation toward Stable and Responsive Microbubbles and Their Use as a Resuscitative Fluid. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201711839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yifeng Peng
- Translational Research Laboratory; Department of Cardiology, Boston Children's Hospital; Department of Pediatrics; Harvard Medical School; Boston MA 02115 USA
| | - Raymond P. Seekell
- Translational Research Laboratory; Department of Cardiology, Boston Children's Hospital; Department of Pediatrics; Harvard Medical School; Boston MA 02115 USA
| | - Alexis R. Cole
- Translational Research Laboratory; Department of Cardiology; Boston Children's Hospital; Boston MA 02115 USA
| | - Jemima R. Lamothe
- Translational Research Laboratory; Department of Cardiology; Boston Children's Hospital; Boston MA 02115 USA
| | - Andrew T. Lock
- Translational Research Laboratory; Department of Cardiology; Boston Children's Hospital; Boston MA 02115 USA
| | - Sarah van den Bosch
- Translational Research Laboratory; Department of Cardiology; Boston Children's Hospital; Boston MA 02115 USA
| | - Xiaoqi Tang
- Translational Research Laboratory; Department of Cardiology; Boston Children's Hospital; Boston MA 02115 USA
| | - John N. Kheir
- Translational Research Laboratory; Department of Cardiology, Boston Children's Hospital; Department of Pediatrics; Harvard Medical School; Boston MA 02115 USA
| | - Brian D. Polizzotti
- Translational Research Laboratory; Department of Cardiology, Boston Children's Hospital; Department of Pediatrics; Harvard Medical School; Boston MA 02115 USA
| |
Collapse
|