Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Theranostics ; 11(19): 9557-9570, 2021.
Article in English | MEDLINE | ID: mdl-34646386

ABSTRACT

Purpose: Preclinical and clinical data indicate that contrast-enhanced ultrasound can enhance tumor perfusion and vessel permeability, thus, improving chemotherapy accumulation and therapeutic outcome. Therefore, we investigated the effects of high mechanical index (MI) contrast-enhanced Doppler ultrasound (CDUS) on tumor perfusion in breast cancer. Methods: In this prospective study, breast cancer patients were randomly assigned to receive either 18 minutes of high MI CDUS during chemotherapy infusion (n = 6) or chemotherapy alone (n = 5). Tumor perfusion was measured before and after at least six chemotherapy cycles using motion-model ultrasound localization microscopy. Additionally, acute effects of CDUS on vessel perfusion and chemotherapy distribution were evaluated in mice bearing triple-negative breast cancer (TNBC). Results: Morphological and functional vascular characteristics of breast cancer in patients were not significantly influenced by high MI CDUS. However, complete clinical tumor response after neoadjuvant chemotherapy was lower in high MI CDUS-treated (1/6) compared to untreated patients (4/5) and size reduction of high MI CDUS treated tumors tended to be delayed at early chemotherapy cycles. In mice with TNBC high MI CDUS decreased the perfused tumor vessel fraction (p < 0.01) without affecting carboplatin accumulation or distribution. Higher vascular immaturity and lower stromal stabilization may explain the stronger vascular response in murine than human tumors. Conclusion: High MI CDUS had no detectable effect on breast cancer vascularization in patients. In mice, the same high MI CDUS setting did not affect chemotherapy accumulation although strong effects on the tumor vasculature were detected histologically. Thus, sonopermeabilization in human breast cancers might not be effective using high MI CDUS protocols and future applications may rather focus on low MI approaches triggering microbubble oscillations instead of destruction. Furthermore, our results show that there are profound differences in the response of mouse and human tumor vasculature to high MI CDUS, which need to be further explored and considered in clinical translation.


Subject(s)
Breast Neoplasms/therapy , Neoadjuvant Therapy/methods , Ultrasonic Therapy/methods , Adult , Animals , Carboplatin/administration & dosage , Carboplatin/therapeutic use , Contrast Media/pharmacology , Female , Humans , Mice , Mice, Inbred BALB C , Microbubbles , Middle Aged , Perfusion , Prospective Studies , Triple Negative Breast Neoplasms/drug therapy , Ultrasonography , Ultrasonography, Doppler/methods
2.
Article in English | MEDLINE | ID: mdl-32167890

ABSTRACT

In clinical applications of super-resolution ultrasound imaging, it is often not possible to achieve a full reconstruction of the microvasculature within a limited measurement time. This makes the comparison of studies and quantitative parameters of vascular morphology and perfusion difficult. Therefore, saturation models were proposed to predict adequate measurement times and estimate the degree of vessel reconstruction. Here, we derive a statistical model for the microbubble counts in super-resolution voxels by a zero-inflated Poisson (ZIP) process. In this model, voxels either belong to vessels with probability Pv and count events with Poisson rate Λ , or they are empty and remain zero. In this model, Pv represents the vessel voxel density in the super-resolution image after infinite measurement time. For the parameters Pv and Λ , we give Cramér-Rao lower bounds (CRLBs) for the estimation variance and derive maximum likelihood estimators (MLEs) in a novel closed-form solution. These can be calculated with knowledge of only the counts at the end of the acquisition time. The estimators are applied to preclinical and clinical data and the MLE outperforms alternative estimators proposed before. The estimated degree of reconstruction lies between 38% and 74% after less than 90 s. Vessel probability Pv ranged from 4% to 20%. The rate parameter Λ was estimated in the range of 0.5-1.3 microbubbles/voxel. For these parameter ranges, the CRLB gives standard deviations of less than 2%, which supports that the parameters can be estimated with good precision already for limited acquisition times.

3.
Article in English | MEDLINE | ID: mdl-30273150

ABSTRACT

Recently, we proved in the first measurements of breast carcinomas the feasibility of super-resolution ultrasound (US) imaging by motion-model ultrasound localization microscopy in a clinical setup. Nevertheless, pronounced in-plane and out-of-plane motions, a nonoptimized microbubble injection scheme, the lower frame rate and the larger slice thickness made the processing more complex than in preclinical investigations. Here, we compare the results of state-of-the-art contrast-enhanced to super-resolution US imaging and systematically analyze the measurements to get indications for the improvement of image acquisition and processing in the future clinical studies. In this regard, the application of a saturation model to the reconstructed vessels is shown to be a valuable tool not only to estimate the measurement times necessary to adequately reconstruct the microvasculature but also for the validation of the measurements. The parameters from this model can also serve to optimize contrast agent concentration and injection protocols. Finally, for the measurements of well-perfused tumors, we observed between 28% and 50% filling for 90-s examination times.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast/diagnostic imaging , Image Interpretation, Computer-Assisted/methods , Ultrasonography, Mammary/methods , Breast Neoplasms/blood supply , Female , Humans , Microbubbles , Microvessels/diagnostic imaging , Pilot Projects
4.
Nat Commun ; 9(1): 1527, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29670096

ABSTRACT

Super-resolution imaging methods promote tissue characterization beyond the spatial resolution limits of the devices and bridge the gap between histopathological analysis and non-invasive imaging. Here, we introduce motion model ultrasound localization microscopy (mULM) as an easily applicable and robust new tool to morphologically and functionally characterize fine vascular networks in tumors at super-resolution. In tumor-bearing mice and for the first time in patients, we demonstrate that within less than 1 min scan time mULM can be realized using conventional preclinical and clinical ultrasound devices. In this context, next to highly detailed images of tumor microvascularization and the reliable quantification of relative blood volume and perfusion, mULM provides multiple new functional and morphological parameters that discriminate tumors with different vascular phenotypes. Furthermore, our initial patient data indicate that mULM can be applied in a clinical ultrasound setting opening avenues for the multiparametric characterization of tumors and the assessment of therapy response.


Subject(s)
Image Processing, Computer-Assisted/methods , Microscopy/methods , Motion , Neoplasms/diagnostic imaging , Neoplasms/pathology , Ultrasonography/methods , A549 Cells , Algorithms , Animals , Cell Line, Tumor , Contrast Media/chemistry , Female , Humans , Mice , Microbubbles , Middle Aged , Neoplasm Transplantation , Phenotype , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL