34 research outputs found

    Commentary on the detection of bubble activity generated in ex-vivo tissue by high intensity focused ultrasound (HIFU) with respect to the generation of therapeutic lesions in tissue for the treatment of cancer

    No full text
    Cancer treatment by extracorporeal high intensity focused ultrasound (HIFU) is constrained by the time needed to ablate relevant tumour volumes clinically. Controlled cavitation might be used to optimise HIFU treatments, but such control requires a greater understanding of its role in lesion formation, and the provision of appropriate techniques to monitor cavitation in tissue. During HIFU exposure various forms of cavitation can occur: acoustic cavitation (both non-inertial and inertial), and bubble formation due to two thermally-driven effects (the vaporisation of liquid into vapour, and the exsolution of formerly dissolved permanent gas out of the liquid and into gas spaces). Different forms of cavitation gives rise to characteristic signals that can be monitored during HIFU. Furthermore, the character of the signal can change depending on the stage of the cavitation in question (nucleation, established cavitation, population effects etc.). Prior to undertaking experiments using tissue, studies were performed by exposing degassed water. The aim of these experiments was to test a detection system in a minimally attenuating medium known to cavitate, in order to provide data for comparison with ex-vivo tissue results.This report is written in support of the journal paper “A Study of Bubble Activity Generated in Ex-Vivo Tissue by High Intensity Focused Ultrasound (HIFU)” by the same authors (McLaughlan et al. 2010). It outlines supporting material for the discussion and conclusions contained in that paper, a study involving monitoring clinically relevant HIFU exposures in degassed water and ex-vivo bovine liver. Monitoring is accomplished using a suite of cavitation detection techniques (exploiting passive and active acoustics, audible and ultrasonic emissions and electrical drive power fluctuations). The aim of the paper is to allow informed progress towards a monitoring system specifically tailored for use during clinical HIFU treatment

    Focused Ultrasound-Mediated Hyperthermia in Vitro: An Experimental Arrangement for Treating Cells under Tissue-Mimicking Conditions.

    Get PDF
    An experimental arrangement that allows in vitro exposure of cells to focused ultrasound-mediated hyperthermia (43°C-55°C) in a tissue-mimicking phantom with biological, acoustic and thermal properties comparable to those of human soft tissue is described. Cells were embedded in a compressed collagen gel, which was sandwiched between 6-mm-thick slices of biocompatible, acoustically absorbing and thermally tissue mimicking poly(vinyl alcohol) cryo-gel. To illustrate the system's potential, cells were exposed using a 1.66-MHz focused ultrasound beam (spatial-peak temporal-average intensities (ISPTA) = 900-1400 W/cm2) that traced out a circular trajectory (5-8 mm in diameter). Real-time temperature monitoring allowed cells to be exposed reproducibly to a pre-determined thermal dose. An experimental planning tool that estimates the thermal dose distribution throughout the sample and allows spatial correlation with cell position has been developed. Treatment response was evaluated qualitatively using microscopy and cell viability testing. This experimental arrangement has significant potential for future, biologically relevant, in vitro focused ultrasound-mediated hyperthermia studies

    A cellular automaton model for spheroid response to radiation and hyperthermia treatments.

    Get PDF
    Thermo-radiosensitisation is a promising approach for treatment of radio-resistant tumours such as those containing hypoxic subregions. Response prediction and treatment planning should account for tumour response heterogeneity, e.g. due to microenvironmental factors, and quantification of the biological effects induced. 3D tumour spheroids provide a physiological in vitro model of tumour response and a systems oncology framework for simulating spheroid response to radiation and hyperthermia is presented. Using a cellular automaton model, 3D oxygen diffusion, delivery of radiation and/or hyperthermia were simulated for many ([Formula: see text]) individual cells forming a spheroid. The iterative oxygen diffusion model was compared to an analytical oxygenation model and simulations were calibrated and validated against experimental data for irradiated (0-10 Gy) and/or heated (0-240 CEM43) HCT116 spheroids. Despite comparable clonogenic survival, spheroid growth differed significantly following radiation or hyperthermia. This dynamic response was described well by the simulation ([Formula: see text] > 0.85). Heat-induced cell death was implemented as a fast, proliferation-independent process, allowing reoxygenation and repopulation, whereas radiation was modelled as proliferation-dependent mitotic catastrophe. This framework stands out both through its experimental validation and its novel ability to predict spheroid response to multimodality treatment. It provides a good description of response where biological dose-weighting based on clonogenic survival alone was insufficient

    3D tumour spheroids for the prediction of the effects of radiation and hyperthermia treatments.

    Get PDF
    For multimodality therapies such as the combination of hyperthermia and radiation, quantification of biological effects is key for dose prescription and response prediction. Tumour spheroids have a microenvironment that more closely resembles that of tumours in vivo and may thus be a superior in vitro cancer model than monolayer cultures. Here, the response of tumour spheroids formed from two established human cancer cell lines (HCT116 and CAL27) to single and combination treatments of radiation (0-20 Gy), and hyperthermia at 47 °C (0-780 CEM43) has been evaluated. Response was analysed in terms of spheroid growth, cell viability and the distribution of live/dead cells. Time-lapse imaging was used to evaluate mechanisms of cell death and cell detachment. It was found that sensitivity to heat in spheroids was significantly less than that seen in monolayer cultures. Spheroids showed different patterns of shrinkage and regrowth when exposed to heat or radiation: heated spheroids shed dead cells within four days of heating and displayed faster growth post-exposure than samples that received radiation or no treatment. Irradiated spheroids maintained a dense structure and exhibited a longer growth delay than spheroids receiving hyperthermia or combination treatment at (thermal) doses that yielded equivalent levels of clonogenic cell survival. We suggest that, unlike radiation, which kills dividing cells, hyperthermia-induced cell death affects cells independent of their proliferation status. This induces microenvironmental changes that promote spheroid growth. In conclusion, 3D tumour spheroid growth studies reveal differences in response to heat and/or radiation that were not apparent in 2D clonogenic assays but that may significantly influence treatment efficacy

    The Feasibility of Thermal Imaging as a Future Portal Imaging Device for Therapeutic Ultrasound.

    Get PDF
    This technical note describes a prototype thermally based portal imaging device that allows mapping of energy deposition on the surface of a tissue mimicking material in a focused ultrasound surgery (FUS) beam by using an infrared camera to measure the temperature change on that surface. The aim of the work is to explore the feasibility of designing and building a system suitable for rapid quality assurance (QA) for use with both ultrasound- and magnetic resonance (MR) imaging-guided clinical therapy ultrasound systems. The prototype was tested using an MR-guided Sonalleve FUS system (with the treatment couch outside the magnet bore). The system's effective thermal noise was 0.02°C, and temperature changes as low as 0.1°C were easily quantifiable. The advantages and drawbacks of thermal imaging for QA are presented through analysis of the results of an experimental session

    Focused ultrasound transducer spatial peak intensity estimation: a comparison of methods

    Get PDF
    Characterisation of the spatial peak intensity at the focus of high intensity focused ultrasound transducers is difficult because of the risk of damage to hydrophone sensors at the high focal pressures generated. Hill et al (1994 Ultrasound Med. Biol. 20 259-69) provided a simple equation for estimating spatial-peak intensity for solid spherical bowl transducers using measured acoustic power and focal beamwidth. This paper demonstrates theoretically and experimentally that this expression is only strictly valid for spherical bowl transducers without a central (imaging) aperture. A hole in the centre of the transducer results in over-estimation of the peak intensity. Improved strategies for determining focal peak intensity from a measurement of total acoustic power are proposed. Four methods are compared: (i) a solid spherical bowl approximation (after Hill et al 1994 Ultrasound Med. Biol. 20 259-69), (ii) a numerical method derived from theory, (iii) a method using measured sidelobe to focal peak pressure ratio, and (iv) a method for measuring the focal power fraction (FPF) experimentally. Spatial-peak intensities were estimated for 8 transducers at three drive powers levels: low (approximately 1 W), moderate (~10 W) and high (20-70 W). The calculated intensities were compared with those derived from focal peak pressure measurements made using a calibrated hydrophone. The FPF measurement method was found to provide focal peak intensity estimates that agreed most closely (within 15%) with the hydrophone measurements, followed by the pressure ratio method (within 20%). The numerical method was found to consistently over-estimate focal peak intensity (+40% on average), however, for transducers with a central hole it was more accurate than using the solid bowl assumption (+70% over-estimation). In conclusion, the ability to make use of an automated beam plotting system, and a hydrophone with good spatial resolution, greatly facilitates characterisation of the FPF, and consequently gives improved confidence in estimating spatial peak intensity from measurement of acoustic power

    Response to comment by G. Borasi.

    Full text link

    'Relationship between thermal dose and cell death for "rapid" ablative and "slow" hyperthermic heating'.

    Get PDF
    Aim Thermal isoeffective dose (TID) has not been convincingly validated for application to predict biological effects from rapid thermal ablation (e.g., using >55 °C). This study compares the classical method of quantifying TID (derived from hyperthermia data) with a temperature-adjusted method based on the Arrhenius model for predicting cell survival in vitro, after either 'rapid' ablative or 'slow' hyperthermic exposures.Methods MTT assay viability data was obtained from two human colon cancer cell lines, (HCT116, HT29), subjected to a range of TIDs (120-720 CEM43) using a thermal cycler for hyperthermic (>2 minutes, 55 °C). TID was initially estimated using a constant RCEM>43°C=0.5, and subsequently using RCEM(T), derived from temperature dependent cell survival (injury rate) Arrhenius analysis.Results 'Slow' and 'rapid' exposures resulted in cell survival and significant regrowth (both cell lines) 10 days post-treatment for 240 CEM43 (RCEM>43°C=0.5), while 340-550 CEM43 (RCEM>43°C =0.5) delivered using 'rapid' exposures showed 12 ± 6% viability and 'slow' exposures resulted in undetectable viability. Arrhenius analysis of experimental data (activation energy ΔE = 5.78 ± 0.04 × 105 J mole-1, frequency factor A = 3.27 ± 11 × 1091 sec-1) yielded RCEM=0.42 * e0.0041*T which better-predicted cell survival than using R CEM> 43°C=0.5.Conclusions TID calculated using an RCEM(T) informed by Arrhenius kinetic parameters provided a more consistent, heating strategy independent, predictor of cell viability, improving dosimetry of ablative thermal exposures. Cell viability was only undetectable above 305 ± 10 CEM43 using this revised measure

    MR guided high intensity focused ultrasound (MRgHIFU) for treating recurrent gynaecological tumours: a pilot feasibility study.

    Get PDF
    Objective To assess the feasibility of targeting recurrent gynaecological tumours with MR guided high intensity focused ultrasound (MRgHIFU).Methods 20 patients with recurrent gynaecological tumours were prospectively scanned on a Philips/Profound 3 T Achieva MR/ Sonalleve HIFU system. Gross tumour volume (GTV) and planning target volume (PTV) were delineated on T 2W and diffusion-weighted imaging (DWI). Achievable treatment volumes that (i) assumed bowel and/or urogenital tract preparation could be used to reduce risk of damage to organs-at-risk (TVoptimal), or (ii) assumed no preparations were possible (TVno-prep) were compared with PTV on virtual treatment plans. Patients were considered treatable if TVoptimal ≥ 50 % PTV.Results 11/20 patients (55%) were treatable if preparation strategies were used: nine had central pelvic recurrences, two had tumours in metastatic locations. Treatable volume ranged from 3.4 to 90.3 ml, representing 70 ± 17 % of PTVs. Without preparation, 6/20 (30%) patients were treatable (four central recurrences, two metastatic lesions). Limiting factors were disease beyond reach of the HIFU transducer, and bone obstructing tumour access. DWI assisted tumour outlining, but differences from T 2W imaging in GTV size (16.9 ± 23.0%) and PTV location (3.8 ± 2.8 mm in phase-encode direction) limited its use for treatment planning.Conclusions Despite variation in size and location within the pelvis, ≥ 50 % of tumour volumes were considered targetable in 55 % patients while avoiding adjacent critical structures. A prospective treatment study will assess safety and symptom relief in a second patient cohort.Advances in knowledge Target size, location and access make MRgHIFU a viable treatment modality for treating symptomatic recurrent gynaecological tumours within the pelvis

    A comprehensive model for heat-induced radio-sensitisation.

    Get PDF
    Combined radiotherapy (RT) and hyperthermia (HT) treatments may improve treatment outcome by heat induced radio-sensitisation. We propose an empirical cell survival model (AlphaR model) to describe this multimodality therapy. The model is motivated by the observation that heat induced radio-sensitisation may be explained by a reduction in the DNA damage repair capacity of heated cells. We assume that this repair is only possible up to a threshold level above which survival will decrease exponentially with dose. Experimental cell survival data from two cell lines (HCT116, Cal27) were considered along with that taken from the literature (baby hamster kidney [BHK] and Chinese hamster ovary cells [CHO]) for HT and combined RT-HT. The AlphaR model was used to study the dependence of clonogenic survival on treatment temperature, and thermal dose R2 ≥ 0.95 for all fits). For HT survival curves (0-80 CEM43 at 43.5-57 °C), the number of free fit AlphaR model parameters could be reduced to two. Both parameters increased exponentially with temperature. We derived the relative biological effectiveness (RBE) or HT treatments at different temperatures, to provide an alternative description of thermal dose, based on our AlphaR model. For combined RT-HT, our analysis is restricted to the linear quadratic arm of the model. We show that, for the range used (20-80 CEM43, 0-12 Gy), thermal dose is a valid indicator of heat induced radio-sensitisation, and that the model parameters can be described as a function thereof. Overall, the proposed model provides a flexible framework for describing cell survival curves, and may contribute to better quantification of heat induced radio-sensitisation, and thermal dose in general
    corecore