Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Ultrasonics ; 137: 107179, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37939413

RESUMO

Ultrasound is an adjunct tool to mammography that can quickly and safely aid physicians in diagnosing breast abnormalities. Clinical ultrasound often assumes a constant sound speed to form diagnostic B-mode images. However, the components of breast tissue, such as glandular tissue, fat, and lesions, differ in sound speed. Given a constant sound speed assumption, these differences can degrade the quality of reconstructed images via phase aberration. Sound speed images can be a powerful tool for improving image quality and identifying diseases if properly estimated. To this end, we propose a supervised deep-learning approach for sound speed estimation from analytic ultrasound signals. We develop a large-scale simulated ultrasound dataset that generates representative breast tissue samples by modeling breast gland, skin, and lesions with varying echogenicity and sound speed. We adopt a fully convolutional neural network architecture trained on a simulated dataset to produce an estimated sound speed map. The simulated tissue is interrogated with a plane wave transmit sequence, and the complex-value reconstructed images are used as input for the convolutional network. The network is trained on the sound speed distribution map of the simulated data, and the trained model can estimate sound speed given reconstructed pulse-echo signals. We further incorporate thermal noise augmentation during training to enhance model robustness to artifacts found in real ultrasound data. To highlight the ability of our model to provide accurate sound speed estimations, we evaluate it on simulated, phantom, and in-vivo breast ultrasound data.


Assuntos
Aprendizado Profundo , Humanos , Feminino , Algoritmos , Ultrassonografia Mamária , Som , Ultrassonografia/métodos , Imagens de Fantasmas , Processamento de Imagem Assistida por Computador/métodos
2.
Int J Mol Sci ; 24(10)2023 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-37240396

RESUMO

The main aim of this study is to synthesize contrast microbubbles (MB) functionalized with engineered protein ligands using a microfluidic device to target breast cancer specific vascular B7-H3 receptor in vivo for diagnostic ultrasound imaging. We used a high-affinity affibody (ABY) selected against human/mouse B7-H3 receptor for engineering targeted MBs (TMBs). We introduced a C-terminal cysteine residue to this ABY ligand for facilitating site-specific conjugation to DSPE-PEG-2K-maleimide (M. Wt = 2.9416 kDa) phospholipid for MB formulation. We optimized the reaction conditions of bioconjugations and applied it for microfluidic based synthesis of TMBs using DSPE-PEG-ABY and DPPC liposomes (5:95 mole %). The binding affinity of TMBs to B7-H3 (MBB7-H3) was tested in vitro in MS1 endothelial cells expressing human B7-H3 (MS1B7-H3) by flow chamber assay, and by ex vivo in the mammary tumors of a transgenic mouse model (FVB/N-Tg (MMTV-PyMT)634Mul/J), expressing murine B7-H3 in the vascular endothelial cells by immunostaining analyses. We successfully optimized the conditions needed for generating TMBs using a microfluidic system. The synthesized MBs showed higher affinity to MS1 cells engineered to express higher level of hB7-H3, and in the endothelial cells of mouse tumor tissue upon injecting TMBs in a live animal. The average number (mean ± SD) of MBB7-H3 binding to MS1B7-H3 cells was estimated to be 354.4 ± 52.3 per field of view (FOV) compared to wild-type control cells (MS1WT; 36.2 ± 7.5/FOV). The non-targeted MBs did not show any selective binding affinity to both the cells (37.7 ± 7.8/FOV for MS1B7-H3 and 28.3 ± 6.7/FOV for MS1WT cells). The fluorescently labeled MBB7-H3 upon systemic injection in vivo co-localized to tumor vessels, expressing B7-H3 receptor, as validated by ex vivo immunofluorescence analyses. We have successfully synthesized a novel MBB7-H3 via microfluidic device, which allows us to produce on demand TMBs for clinical applications. This clinically translatable MBB7-H3 showed significant binding affinity to vascular endothelial cells expressing B7-H3 both in vitro and in vivo, which shows its potential for clinical translation as a molecular ultrasound contrast agent for human applications.


Assuntos
Neoplasias da Mama , Receptores Histamínicos H3 , Camundongos , Animais , Humanos , Feminino , Microbolhas , Células Endoteliais/metabolismo , Ultrassonografia/métodos , Camundongos Transgênicos , Imagem Molecular/métodos , Meios de Contraste , Neoplasias da Mama/patologia , Dispositivos Lab-On-A-Chip
5.
J Exp Clin Cancer Res ; 41(1): 299, 2022 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-36224614

RESUMO

BACKGROUND: Therapeutic strategies engaging the immune system against malignant cells have revolutionized the field of oncology. Proficiency of dendritic cells (DCs) for antigen presentation and immune response has spurred interest on DC-based vaccines for anti-cancer therapy. However, despite favorable safety profiles in patients, current DC-vaccines have not yet presented significant outcome due to technical barriers in active DC delivery, tumor progression, and immune dysfunction. To maximize the therapeutic response, we present here a unique cell-free DC-based vaccine capable of lymphoid organ targeting and eliciting T-cell-mediated anti-tumor effect. METHODS: We developed this novel immunotheranostic platform using plasma membranes derived from activated DCs incorporated into ultrasound contrast microbubbles (MBs), thereby offering real-time visualization of MBs' trafficking and homing in vivo. Human PBMC-derived DCs were cultured ex vivo for controlled maturation and activation using cell membrane antigens from breast cancer cells. Following DC membrane isolation, immunotheranostic microbubbles, called DC-iMBs, were formed for triple negative breast cancer treatment in a mouse model harboring a human reconstituted immune system. RESULTS: Our results demonstrated that DC-iMBs can accumulate in lymphoid organs and induce anti-tumor immune response, which significantly reduced tumor growth via apoptosis while increasing survival length of the treated animals. The phenotypic changes in immune cell populations upon DC-iMBs delivery further confirmed the T-cell-mediated anti-tumor effect. CONCLUSION: These early findings strongly support the potential of DC-iMBs as a novel immunotherapeutic cell-free vaccine for anti-cancer therapy.


Assuntos
Neoplasias da Mama , Vacinas Anticâncer , Animais , Neoplasias da Mama/tratamento farmacológico , Células Dendríticas , Feminino , Humanos , Imunoterapia/métodos , Leucócitos Mononucleares , Camundongos , Microbolhas
6.
J Nanobiotechnology ; 20(1): 267, 2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35689262

RESUMO

Triple-negative breast cancer (TNBC) is a highly heterogeneous breast cancer subtype with poor prognosis. Although anatomical imaging figures prominently for breast lesion screening, TNBC is often misdiagnosed, thus hindering early medical care. Ultrasound (US) molecular imaging using nanobubbles (NBs) capable of targeting tumor cells holds great promise for improved diagnosis and therapy. However, the lack of conventional biomarkers in TNBC impairs the development of current targeted agents. Here, we exploited the homotypic recognition of cancer cells to synthesize the first NBs based on TNBC cancer cell membrane (i.e., NBCCM) as a targeted diagnostic agent. We developed a microfluidic technology to synthesize NBCCM based on the self-assembly property of cell membranes in aqueous solutions. In vitro, optimal NBCCM had a hydrodynamic diameter of 683 ± 162 nm, showed long-lasting US contrast enhancements and homotypic affinity. In vivo, we demonstrated that NBCCM showed increased extravasation and retention in a TNBC mouse model compared to non-targeted NBs by US molecular imaging. Peak intensities and areas under the curves from time-intensity plots showed a significantly enhanced signal from NBCCM compared to non-targeted NBs (2.1-fold, P = 0.004, and, 3.6-fold, P = 0.0009, respectively). Immunofluorescence analysis further validated the presence of NBCCM in the tumor microenvironment. Circumventing the challenge for universal cancer biomarker identification, our approach could enable TNBC targeting regardless of tumor tissue heterogeneity, thus improving diagnosis and potentially gene/drug targeted delivery. Ultimately, our approach could be used to image many cancer types using biomimetic NBs prepared from their respective cancer cell membranes.


Assuntos
Neoplasias de Mama Triplo Negativas , Animais , Biomimética , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , Imagem Molecular/métodos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Microambiente Tumoral
7.
Nanotheranostics ; 6(1): 62-78, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34976581

RESUMO

Rationale: To assess treatment effects of 4 complementary miRNAs (miRNA-100/miRNA-122/antimiRNA-10b/antimiRNA-21) encapsulated in a biodegradable PLGA-PEG nanoparticle, administered by an ultrasound-guided microbubble-mediated targeted delivery (UGMMTD) approach in mouse models of hepatocellular carcinoma (HCC). Methods:In vitro apoptotic index was measured in HepG2 and Hepa1-6 HCC cells treated with various combinations of the 4 miRNAs with doxorubicin. Three promising combinations were further tested in vivo by using UGMMTD. 63 HepG2 xenografts in mice were randomized into: group 1, miRNA-122/antimiRNA-10b/antimiRNA-21/US/doxorubicin; group 2, miRNA-100/miRNA-122/antimiRNA-10b/antimiRNA-21/US/doxorubicin; group 3, miRNA-100/miRNA-122/antimiRNA-10b/US/doxorubicin; group 4, miRNA-122/anitmiRNA-10b/antimiRNA-21/doxorubicin; group 5, miRNA-100/miRNA-122/antimiRNA-10b/antimiRNA-21/doxorubicin; group 6, miRNA-100/miRNA-122/antimiRNA-10b/doxorubicin; group 7, doxorubicin only treatment; and group 8, without any treatment. Tumor volumes were measured through 18 days. H&E staining, TUNEL assay, and qRT-PCR quantification for delivered miRNAs were performed. Results:In vivo results showed that UGMMTD of miRNAs with doxorubicin in groups 1-3 significantly (P<0.05) delayed tumor growth compared to control without any treatment, and doxorubicin only from day 7 to 18. On qRT-PCR, levels of delivered miRNAs were significantly (P<0.05) higher in groups 1-3 upon UGMMTD treatment compared to controls. TUNEL assay showed that upon UGMMTD, significantly higher levels of apoptotic cell populations were observed in groups 1-3 compared to controls. Toxicity was not observed in various organs of different groups. Conclusions: UGMMTD of miRNA-100/miRNA-122/antimiRNA-10b/antimiRNA-21 combination improved therapeutic outcome of doxorubicin chemotherapy in mouse models of HCC by substantial inhibition of tumor growth and significant increase in apoptotic index.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs , Animais , Humanos , Camundongos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Microbolhas , MicroRNAs/genética , Ultrassonografia de Intervenção
8.
IEEE Trans Med Imaging ; 40(7): 1888-1897, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33755561

RESUMO

Photoacoustic (PA) imaging can revolutionize medical ultrasound by augmenting it with molecular information. However, clinical translation of PA imaging remains a challenge due to the limited viewing angles and imaging depth. Described here is a new robust algorithm called Superiorized Photo-Acoustic Non-NEgative Reconstruction (SPANNER), designed to reconstruct PA images in real-time and to address the artifacts associated with limited viewing angles and imaging depth. The method utilizes precise forward modeling of the PA propagation and reception of signals while accounting for the effects of acoustic absorption, element size, shape, and sensitivity, as well as the transducer's impulse response and directivity pattern. A fast superiorized conjugate gradient algorithm is used for inversion. SPANNER is compared to three reconstruction algorithms: delay-and-sum (DAS), universal back-projection (UBP), and model-based reconstruction (MBR). All four algorithms are applied to both simulations and experimental data acquired from tissue-mimicking phantoms, ex vivo tissue samples, and in vivo imaging of the prostates in patients. Simulations and phantom experiments highlight the ability of SPANNER to improve contrast to background ratio by up to 20 dB compared to all other algorithms, as well as a 3-fold increase in axial resolution compared to DAS and UBP. Applying SPANNER on contrast-enhanced PA images acquired from prostate cancer patients yielded a statistically significant difference before and after contrast agent administration, while the other three image reconstruction methods did not, thus highlighting SPANNER's performance in differentiating intrinsic from extrinsic PA signals and its ability to quantify PA signals from the contrast agent more accurately.


Assuntos
Técnicas Fotoacústicas , Acústica , Algoritmos , Artefatos , Humanos , Processamento de Imagem Assistida por Computador , Imagens de Fantasmas
9.
Artigo em Inglês | MEDLINE | ID: mdl-33226938

RESUMO

Transcranial magnetic resonance-guided focused ultrasound (tcMRgFUS) is gaining significant acceptance as a noninvasive treatment for motion disorders and shows promise for novel applications such as blood-brain barrier opening for tumor treatment. A typical procedure relies on CT-derived acoustic property maps to simulate the transfer of ultrasound through the skull. Accurate estimates of the acoustic attenuation in the skull are essential to accurate simulations, but there is no consensus about how attenuation should be estimated from CT images and there is interest in exploring MR as a predictor of attenuation in the skull. In this study, we measure the acoustic attenuation at 0.5, 1, and 2.25 MHz in 89 samples taken from two ex vivo human skulls. CT scans acquired with a variety of X-ray energies, reconstruction kernels, and reconstruction algorithms, and MR images acquired with ultrashort and zero echo time sequences are used to estimate the average Hounsfield unit value, MR magnitude, and T2* value in each sample. The measurements are used to develop a model of attenuation as a function of frequency and each individual imaging parameter.


Assuntos
Imageamento por Ressonância Magnética , Tomografia Computadorizada por Raios X , Acústica , Algoritmos , Humanos , Crânio/diagnóstico por imagem
10.
Artigo em Inglês | MEDLINE | ID: mdl-33141666

RESUMO

Passive cavitation mapping (PCM) techniques typically utilize a time-exposure acoustic (TEA) approach, where the received radio frequency data are beamformed, squared, and integrated over time. Such PCM-TEA cavitation maps typically suffer from long-tail artifacts and poor axial resolution with pulse-echo diagnostic arrays. Here, we utilize a recently developed PCM technique based on cavitation source localization (CSL), which fits a hyperbolic function to the received cavitation wavefront. A filtering method based on the root-mean-square error (rmse) of the hyperbolic fit is utilized to filter out spurious signals. We apply a wavefront correction technique to the signals with poor fit quality to recover additional cavitation signals and improve cavitation localization. Validation of the PCM-CSL technique with rmse filtering and wavefront correction was conducted in experiments with a tissue-mimicking flow phantom and an in vivo mouse model of cancer. It is shown that the quality of the hyperbolic fit, necessary for the PCM-CSL, requires an rmse < 0.05 mm2 in order to accurately localize the cavitation sources. A detailed study of the wavefront correction technique was carried out, and it was shown that, when applied to experiments with high noise and interference from multiple cavitating microbubbles, it was capable of effectively correcting noisy wavefronts without introducing spurious cavitation sources, thereby improving the quality of the PCM-CSL images. In phantom experiments, the PCM-CSL was capable of precisely localizing sources on the therapy beam axis and off-axis sources. In vivo cavitation experiments showed that PMC-CSL showed a significant improvement over PCM-TEA and yielded acceptable localization of cavitation signals in mice.


Assuntos
Microbolhas , Neoplasias , Acústica , Animais , Artefatos , Camundongos , Imagens de Fantasmas
11.
Ultrasound Med Biol ; 47(2): 309-322, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33153807

RESUMO

Ultrasound and microbubble (USMB)-mediated drug delivery is a valuable tool for increasing the efficiency of the delivery of therapeutic agents to cancer while maintaining low systemic toxicity. Typically, selection of USMB drug delivery parameters used in current research settings are either based on previous studies described in the literature or optimized using tissue-mimicking phantoms. However, phantoms rarely mimic in vivo tumor environments, and the selection of parameters should be based on the application or experiment. In the following study, we optimized the therapeutic parameters of the ultrasound drug delivery system to achieve the most efficient in vivo drug delivery using fluorescent semiconducting polymer nanoparticles as a model nanocarrier. We illustrate that voltage, pulse repetition frequency and treatment time (i.e., number of ultrasound pulses per therapy area) delivered to the tumor can successfully be optimized in vivo to ensure effective delivery of the semiconducting polymer nanoparticles to models of hepatocellular carcinoma. The optimal in vivo parameters for USMB drug delivery in this study were 70 V (peak negative pressure = 3.4 MPa, mechanical index = 1.22), 1-Hz pulse repetition frequency and 100-s therapy time. USMB-mediated drug delivery using in vivo optimized ultrasound parameters caused an up to 2.2-fold (p < 0.01) increase in drug delivery to solid tumors compared with that using phantom-optimized ultrasound parameters.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Neoplasias Hepáticas/tratamento farmacológico , Microbolhas/uso terapêutico , Ondas Ultrassônicas , Animais , Calibragem , Carcinoma Hepatocelular/patologia , Modelos Animais de Doenças , Feminino , Fluorescência , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Necrose , Transplante de Neoplasias , Pontos Quânticos , Terapia por Ultrassom/instrumentação
12.
Cell Transplant ; 29: 963689720965478, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33028105

RESUMO

We studied the paracrine function of mesenchymal stem cells (MSCs) derived from various sources in response to pulsed focused ultrasound (pFUS). Human adipose tissue (AD), bone marrow (BM), and umbilical cord (UC) derived MSCs were exposed to pFUS at two intensities: 0.45 W/cm2 ISATA (310 kPa PNP) and 1.3 W/cm2 ISATA (540 kPa PNP). Following pFUS, the viability and proliferation of MSCs were assessed using a hemocytometer and confocal microscopy, and their secreted cytokine profile determined using a multiplex ELISA. Our findings showed that pFUS can stimulate the production of immunomodulatory, anti-inflammatory, and angiogenic cytokines from MSCs which was dependent on both the source of MSC being studied and the acoustic intensity employed. These important findings set the foundation for additional mechanistic and validation studies using this novel noninvasive and clinically translatable technology for modulating MSC biology.


Assuntos
Células da Medula Óssea/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Ensaio de Imunoadsorção Enzimática , Humanos , Microscopia Confocal
13.
Stem Cell Res Ther ; 11(1): 405, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32948247

RESUMO

BACKGROUND: The aim of this study was to examine the effect of a three-step approach that utilizes the application of adipose tissue-derived mesenchymal stem cells (AD-MSCs), encapsulation, and pulsed focused ultrasound (pFUS) to help the engraftment and function of transplanted islets. METHODS: In step 1, islets were co-cultured with AD-MSCs to form a coating of AD-MSCs on islets: here, AD-MSCs had a cytoprotective effect on islets; in step 2, islets coated with AD-MSCs were conformally encapsulated in a thin layer of alginate using a co-axial air-flow method: here, the capsule enabled AD-MSCs to be in close proximity to islets; in step 3, encapsulated islets coated with AD-MSCs were treated with pFUS: here, pFUS enhanced the secretion of insulin from islets as well as stimulated the cytoprotective effect of AD-MSCs. RESULTS: Our approach was shown to prevent islet death and preserve islet functionality in vitro. When 175 syngeneic encapsulated islets coated with AD-MSCs were transplanted beneath the kidney capsule of diabetic mice, and then followed every 3 days with pFUS treatment until day 12 post-transplantation, we saw a significant improvement in islet function with diabetic animals re-establishing glycemic control over the course of our study (i.e., 30 days). In addition, our approach was able to enhance islet engraftment by facilitating their revascularization and reducing inflammation. CONCLUSIONS: This study demonstrates that our clinically translatable three-step approach is able to improve the function and viability of transplanted islets.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Diabetes Mellitus Experimental/terapia , Camundongos
14.
J Control Release ; 326: 75-90, 2020 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-32554041

RESUMO

Beyond the emerging field of oncological ultrasound molecular imaging, the recent significant advancements in ultrasound and contrast agent technology have paved the way for therapeutic ultrasound mediated microbubble oscillation and has shown that this approach is capable of increasing the permeability of microvessel walls while also initiating enhanced extravasation and drug delivery into target tissues. In addition, a large number of preclinical studies have demonstrated that ultrasound alone or combined with microbubbles can efficiently increase cell membrane permeability resulting in enhanced tissue distribution and intracellular drug delivery of molecules, nanoparticles, and other therapeutic agents. The mechanism behind the enhanced permeability is the temporary creation of pores in cell membranes through a phenomenon called sonoporation by high-intensity ultrasound and microbubbles or cavitation agents. At low ultrasound intensities (0.3-3 W/cm2), sonoporation may be caused by microbubbles oscillating in a stable motion, also known as stable cavitation. In contrast, at higher ultrasound intensities (greater than 3 W/cm2), sonoporation usually occurs through inertial cavitation that accompanies explosive growth and collapse of the microbubbles. Sonoporation has been shown to be a highly effective method to improve drug uptake through microbubble potentiated enhancement of microvascular permeability. In this review, the therapeutic strategy of using ultrasound for improved drug delivery are summarized with the special focus on cancer therapy. Additionally, we discuss the progress, challenges, and future of ultrasound-mediated drug delivery towards clinical translation.


Assuntos
Microbolhas , Nanopartículas , Permeabilidade da Membrana Celular , Sistemas de Liberação de Medicamentos , Ultrassonografia
15.
IEEE Trans Med Imaging ; 39(10): 3079-3088, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32286963

RESUMO

Ultrasound molecular imaging (UMI) is enabled by targeted microbubbles (MBs), which are highly reflective ultrasound contrast agents that bind to specific biomarkers. Distinguishing between adherent MBs and background signals can be challenging in vivo. The preferred preclinical technique is differential targeted enhancement (DTE), wherein a strong acoustic pulse is used to destroy MBs to verify their locations. However, DTE intrinsically cannot be used for real-time imaging and may cause undesirable bioeffects. In this work, we propose a simple 4-layer convolutional neural network to nondestructively detect adherent MB signatures. We investigated several types of input data to the network: "anatomy-mode" (fundamental frequency), "contrast-mode" (pulse-inversion harmonic frequency), or both, i.e., "dual-mode", using IQ channel signals, the channel sum, or the channel sum magnitude. Training and evaluation were performed on in vivo mouse tumor data and microvessel phantoms. The dual-mode channel signals yielded optimal performance, achieving a soft Dice coefficient of 0.45 and AUC of 0.91 in two test images. In a volumetric acquisition, the network best detected a breast cancer tumor, resulting in a generalized contrast-to-noise ratio (GCNR) of 0.93 and Kolmogorov-Smirnov statistic (KSS) of 0.86, outperforming both regular contrast mode imaging (GCNR = 0.76, KSS = 0.53) and DTE imaging (GCNR = 0.81, KSS = 0.62). Further development of the methodology is necessary to distinguish free from adherent MBs. These results demonstrate that neural networks can be trained to detect targeted MBs with DTE-like quality using nondestructive dual-mode data, and can be used to facilitate the safe and real-time translation of UMI to clinical applications.


Assuntos
Aprendizado Profundo , Microbolhas , Animais , Meios de Contraste , Humanos , Camundongos , Imagem Molecular , Ultrassonografia
16.
Stem Cells Transl Med ; 9(8): 850-866, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32157802

RESUMO

Mesenchymal stromal cells (MSCs) have been a popular platform for cell-based therapy in regenerative medicine due to their propensity to home to damaged tissue and act as a repository of regenerative molecules that can promote tissue repair and exert immunomodulatory effects. Accordingly, a great deal of research has gone into optimizing MSC homing and increasing their secretion of therapeutic molecules. A variety of methods have been used to these ends, but one emerging technique gaining significant interest is the use of ultrasound. Sound waves exert mechanical pressure on cells, activating mechano-transduction pathways and altering gene expression. Ultrasound has been applied both to cultured MSCs to modulate self-renewal and differentiation, and to tissues-of-interest to make them a more attractive target for MSC homing. Here, we review the various applications of ultrasound to MSC-based therapies, including low-intensity pulsed ultrasound, pulsed focused ultrasound, and extracorporeal shockwave therapy, as well as the use of adjunctive therapies such as microbubbles. At a molecular level, it seems that ultrasound transiently generates a local gradient of cytokines, growth factors, and adhesion molecules that facilitate MSC homing. However, the molecular mechanisms underlying these methods are far from fully elucidated and may differ depending on the ultrasound parameters. We thus put forth minimal criteria for ultrasound parameter reporting, in order to ensure reproducibility of studies in the field. A deeper understanding of these mechanisms will enhance our ability to optimize this promising therapy to assist MSC-based approaches in regenerative medicine.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Mesenquimais/metabolismo , Ultrassonografia/métodos , Diferenciação Celular , Humanos , Células-Tronco Mesenquimais/citologia
17.
Clin Cancer Res ; 26(9): 2140-2150, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31924738

RESUMO

PURPOSE: Human B7-H3 (hB7-H3) is a promising molecular imaging target differentially expressed on the neovasculature of breast cancer and has been validated for preclinical ultrasound (US) imaging with anti-B7-H3-antibody-functionalized microbubbles (MB). However, smaller ligands such as affibodies (ABY) are more suitable for the design of clinical-grade targeted MB. EXPERIMENTAL DESIGN: Binding of ABYB7-H3 was confirmed with soluble and cell-surface B7-H3 by flow cytometry. MB were functionalized with ABYB7-H3 or anti-B7-H3-antibody (AbB7-H3). Control and targeted MB were tested for binding to hB7-H3-expressing cells (MS1hB7-H3) under shear stress conditions. US imaging was performed with MBABY-B7-H3 in an orthotopic mouse model of human MDA-MB-231 coimplanted with MS1hB7-H3 or control MS1WT cells and a transgenic mouse model of breast cancer development. RESULTS: ABYB7-H3 specifically binds to MS1hB7-H3 and murine-B7-H3-expressing monocytes. MBABY-B7-H3 (8.5 ± 1.4 MB/cell) and MBAb-B7-H3 (9.8 ± 1.3 MB/cell) showed significantly higher (P < 0.0001) binding to the MS1hB7-H3 cells compared with control MBNon-targeted (0.5 ± 0.1 MB/cell) under shear stress conditions. In vivo, MBABY-B7-H3 produced significantly higher (P < 0.04) imaging signal in orthotopic tumors coengrafted with MS1hB7-H3 (8.4 ± 3.3 a.u.) compared with tumors with MS1WT cells (1.4 ± 1.0 a.u.). In the transgenic mouse tumors, MBABY-B7-H3 (9.6 ± 2.0 a.u.) produced higher (P < 0.0002) imaging signal compared with MBNon-targeted (1.3 ± 0.3 a.u.), whereas MBABY-B7-H3 signal in normal mammary glands and tumors with B7-H3 blocking significantly reduced (P < 0.02) imaging signal. CONCLUSIONS: MBABY-B7-H3 enhances B7-H3 molecular signal in breast tumors, improving cancer detection, while offering the advantages of a small size ligand and easier production for clinical imaging.


Assuntos
Antígenos B7/metabolismo , Neoplasias da Mama/irrigação sanguínea , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Antígenos B7/imunologia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Meios de Contraste/química , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Microbolhas , Imagem Molecular/métodos , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ultrassonografia/métodos
18.
Ultrasound Med Biol ; 46(3): 630-638, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31882169

RESUMO

Pulsed focused ultrasound (pFUS) utilizes short cycles of sound waves to mechanically shake cells within tissues which, in turn, causes transient local increases in cytokines, growth factors and cell adhesion molecules. Although the effect of pFUS has been investigated in several different organs including the kidney, muscle and heart, its effect on the pancreas has not been investigated. In the present work, we applied pFUS to the rodent pancreas with the following parameters: 1.1-MHz frequency, 5-Hz pulse repetition frequency, 5% duty cycle, 10-ms pulse length, 160-s duration. Low-intensity pFUS had a spatial average temporal average intensity of 11.5 W/cm2 and a negative peak pressure of 3 MPa; high-intensity pFUS had a spatial average temporal average intensity of 18.5 W/cm2 and negative peak pressure of 4 MPa. Here we found that pFUS changed the expression of several cytokines while having no effect on the underlying tissue histology or health of pancreatic cells (as reflected by no significant change in plasma levels of amylase and lipase). Furthermore, we found that this effect on cytokine expression in the pancreas was acoustic intensity dependent; while pFUS at low intensities turned off the expression of several cytokines, at high intensities it had the opposite effect and turned on the expression of these cytokines. The ability to non-invasively manipulate the microenvironment of the pancreas using sound waves could have profound implications for priming and modulating this organ for the application of cellular therapies in the context of both regenerative medicine (i.e., diabetes and pancreatitis) and oncology (i.e., pancreatic cancer).


Assuntos
Pâncreas/efeitos da radiação , Ondas Ultrassônicas , Animais , Feminino , Camundongos , Pâncreas/patologia
19.
Adv Ther (Weinh) ; 3(12)2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33415184

RESUMO

Spatially localized microbubble cavitation by ultrasound offers an effective means of altering permeability of natural barriers (i.e. blood vessel and cell membrane) in favor of nanomaterials accumulation in the target site. In this study, a clinically relevant, minimally invasive ultrasound guided therapeutic approach is investigated for targeted delivery of anticancer microRNA loaded PLGA-b-PEG nanoparticles to spontaneous hepatocellular neoplasia in a canine model. Quantitative assessment of the delivered microRNAs revealed prominent and consistent increase in miRNAs levels (1.5-to 2.3-fold increase (p<0.001)) in ultrasound treated tumor regions compared to untreated control regions. Immunohistology of ultrasound treated tumor tissue presented a clear evidence for higher amount of nanoparticles extravasation from the blood vessels. A distinct pattern of cytokine expression supporting CD8+ T cells mediated "cold-to-hot" tumor transition was evident in all patients. On the outset, proposed platform can enhance delivery of miRNA-loaded nanoparticles to deep seated tumors in large animals to enhance chemotherapy.

20.
J Control Release ; 309: 1-10, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31326463

RESUMO

In this study, we designed and validated a platform for ultrasound and microbubble-mediated delivery of FDA-approved pegylated poly lactic-co-glycolic acid (PLGA) nanoparticles loaded with anticancer microRNAs (miRNAs) to deep tissues in a pig model. Small RNAs have been shown to reprogram tumor cells and sensitize them to clinically used chemotherapy. To overcome their short intravascular circulation half-life and achieve controlled and sustained release into tumor cells, anticancer miRNAs need to be encapsulated into nanocarriers. Focused ultrasound combined with gas-filled microbubbles provides a noninvasive way to improve the permeability of tumor vasculature and increase the delivery efficiency of drug-loaded particles. A single handheld, curvilinear ultrasound array was used in this study for image-guided therapy with clinical-grade SonoVue contrast agent. First, we validated the platform on phantoms to optimize the microbubble cavitation dose based on acoustic parameters, including peak negative pressure, pulse length, and pulse repetition frequency. We then tested the system in vivo by delivering PLGA nanoparticles co-loaded with antisense-miRNA-21 and antisense-miRNA-10b to pig liver and kidney. Enhanced miRNA delivery was observed (1.9- to 3.7-fold increase) as a result of the ultrasound treatment compared to untreated control regions. Additionally, we used highly fluorescent semiconducting polymer nanoparticles to visually assess nanoparticle extravasation. Fluorescent microscopy suggested the presence of nanoparticles in the extravascular compartment. Hematoxylin and eosin staining of treated tissues did not reveal tissue damage. The results presented in this manuscript suggest that the proposed platform may be used to safely and noninvasively enhance the delivery of miRNA-loaded nanoparticles to target regions in deep organs in large animal models.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Nanopartículas/química , Neoplasias/terapia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , RNA Antissenso/administração & dosagem , Animais , Sistemas de Liberação de Medicamentos/métodos , Feminino , Terapia Genética , MicroRNAs/genética , Microbolhas , Neoplasias/genética , RNA Antissenso/genética , RNA Antissenso/farmacocinética , Suínos , Terapia por Ultrassom/instrumentação , Terapia por Ultrassom/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA