121 research outputs found

    Pixel-Based Absorption Correction for Dual-Tracer Fluorescence Imaging of Receptor Binding Potential

    Get PDF
    Ratiometric approaches to quantifying molecular concentrations have been used for decades in microscopy, but have rarely been exploited in vivo until recently. One dual-tracer approach can utilize an untargeted reference tracer to account for non-specific uptake of a receptor-targeted tracer, and ultimately estimate receptor binding potential quantitatively. However, interpretation of the relative dynamic distribution kinetics is confounded by differences in local tissue absorption at the wavelengths used for each tracer. This study simulated the influence of absorption on fluorescence emission intensity and depth sensitivity at typical near-infrared fluorophore wavelength bands near 700 and 800 nm in mouse skin in order to correct for these tissue optical differences in signal detection. Changes in blood volume [1-3%] and hemoglobin oxygen saturation [0-100%] were demonstrated to introduce substantial distortions to receptor binding estimates (error \u3e 30%), whereas sampled depth was relatively insensitive to wavelength (error \u3c 6%). In response, a pixel-by-pixel normalization of tracer inputs immediately post-injection was found to account for spatial heterogeneities in local absorption properties. Application of the pixel-based normalization method to an in vivo imaging study demonstrated significant improvement, as compared with a reference tissue normalization approach

    Review of Fluorescence Guided Surgery Systems: Identification of Key Performance Capabilities Beyond Indocyanine Green Imaging

    Get PDF
    There is growing interest in using fluorescence imaging instruments to guide surgery, and the leading options for open-field imaging are reviewed here. While the clinical fluorescence-guided surgery (FGS) field has been focused predominantly on indocyanine green (ICG) imaging, there is accelerated development of more specific molecular tracers. These agents should help advance new indications for which FGS presents a paradigm shift in how molecular information is provided for resection decisions. There has been a steady growth in commercially marketed FGS systems, each with their own differentiated performance characteristics and specifications. A set of desirable criteria is presented to guide the evaluation of instruments, including: (i) real-time overlay of white-light and fluorescence images, (ii) operation within ambient room lighting, (iii) nanomolar-level sensitivity, (iv) quantitative capabilities, (v) simultaneous multiple fluorophore imaging, and (vi) ergonomic utility for open surgery. In this review, United States Food and Drug Administration 510(k) cleared commercial systems and some leading premarket FGS research systems were evaluated to illustrate the continual increase in this performance feature base. Generally, the systems designed for ICG-only imaging have sufficient sensitivity to ICG, but a fraction of the other desired features listed above, with both lower sensitivity and dynamic range. In comparison, the emerging research systems targeted for use with molecular agents have unique capabilities that will be essential for successful clinical imaging studies with low-concentration agents or where superior rejection of ambient light is needed. There is no perfect imaging system, but the feature differences among them are important differentiators in their utility, as outlined in the data and tables here

    Optical Tracer Size Differences Allow Quantitation of Active Pumping Rate Versus Stokes–Einstein Diffusion in Lymphatic Transport

    Get PDF
    Lymphatic uptake of interstitially administered agents occurs by passive convective–diffusive inflow driven by interstitial concentration and pressure, while the downstream lymphatic transport is facilitated by active propulsive contractions of lymphatic vessel walls. Near-infrared fluorescence imaging in mice was used to measure these central components of lymphatic transport for the first time, using two different-sized molecules––methylene blue (MB) and fluorescence-labeled antibody immunoglobulin G (IgG)-IRDye 680RD. This work confirms the hypothesis that lymphatic passive inflow and active propulsion rates can be separated based upon the relative differences in Stokes–Einstein diffusion coefficient. This coefficient specifically affects the passive-diffusive uptake when the interstitial volume and pressure are constant. Parameters such as mean time-to-peak signal, overall fluorescence signal intensities, and number of active peristaltic pulses, were estimated from temporal imaging data. While the mean time to attain peak signal representative of diffusion-dominated flow in the lymph vessels was 0.6±0.2  min for MB and 8±6  min for IgG, showing a size dependence, the active propulsion rates were 3.4±0.8  pulses/min and 3.3±0.5  pulses/min, respectively, appearing size independent. The propulsion rates for both dyes decreased with clearance from the interstitial injection-site, indicating intrinsic control of the smooth muscles in response to interstitial pressure. This approach to size-comparative agent flow imaging of lymphatic function can enable noninvasive characterization of diseases related to uptake and flow in lymph networks

    Fluorescent Affibody Peptide Penetration in Glioma Margin Is Superior to Full Antibody

    Get PDF
    Object: Fluorescence imaging has the potential to significantly improve neurosurgical resection of oncologic lesions through improved differentiation between normal and cancerous tissue at the tumor margins. In order to successfully mark glioma tissue a fluorescent tracer must have the ability to penetrate through the blood brain barrier (BBB) and provide delineation in the tumor periphery where heterogeneously intact BBB may exist. In this study it was hypothesized that, due to its smaller size, fluorescently labeled anti-EGFR Affibody protein (~7 kDa) would provide a more clear delineation of the tumor margin than would fluorescently labeled cetuximab, a full antibody (~150 kDa) to the epidermal growth factor receptor (EGFR). Methods: Cetuximab and anti-EGFR targeted Affibody were conjugated to two different fluorescent dyes (both emitting in the near-infrared) and injected intravenously into 6 athymic mice which were inoculated orthotopically with green fluorescent protein (GFP) expressing human U251 glioma cells. Each mouse was sacrificed at 1-h post injection, at which time brains were removed, snap frozen, sectioned and quantitatively analyzed for fluorescence distribution. Results: Ex vivo analysis showed on average, nearly equal concentrations of cetuximab and Affibody within the tumor (on average Affibody made up 49 ± 6% of injected protein), however, the cetuximab was more confined to the center of the tumor with Affibody showing significantly higher concentrations at the tumor periphery (on average Affibody made up 72 ± 15% of injected protein in the outer 50 um of the tumor). Further ex vivo analysis of detection studies showed that the Affibody provided superior discrimination for differentiation of tumor from surrounding normal brain. Conclusions: The present study indicates that fluorescently labeled anti-EGFR Affibody can provide significantly better delineation of tumor margins than a fluorescently labeled anti-EGFR antibody and shows considerable potential for guiding margin detection during neurosurgery

    Contrast Enhanced-Magnetic Resonance Imaging as a Surrogate to Map Verteporfin Delivery in Photodynamic Therapy

    Get PDF
    The use of in vivo contrast-enhanced magnetic resonance (MR) imaging as a surrogate for photosensitizer (verteporfin) dosimetry in photodynamic therapy of pancreas cancer is demonstrated by correlating MR contrast uptake to ex vivo fluorescence images on excised tissue. An orthotopic pancreatic xenograft mouse model was used for the study. A strong correlation ([i]r=0.57 ) was found for bulk intensity measurements of T1-weighted gadolinium enhancement and verteporfin fluorescence in the tumor region of interest. The use of contrast-enhanced MR imaging shows promise as a method for treatment planning and photosensitizer dosimetry in human photodynamic therapy (PDT) of pancreas cancer

    Improved Tumor Contrast Achieved by Single Time Point Dual-Reporter Fluorescence Imaging

    Get PDF
    In this study, we demonstrate a method to quantify biomarker expression that uses an exogenous dual-reporter imaging approach to improve tumor signal detection. The uptake of two fluorophores, one nonspecific and one targeted to the epidermal growth factor receptor (EGFR), were imaged at 1 h in three types of xenograft tumors spanning a range of EGFR expression levels (n  =  6 in each group). Using this dual-reporter imaging methodology, tumor contrast-to-noise ratio was amplified by \u3e6 times at 1 h postinjection and \u3e2 times at 24 h. Furthermore, by as early as 20 min postinjection, the dual-reporter imaging signal in the tumor correlated significantly with a validated marker of receptor density (P  \u3c  0.05, r  =  0.93). Dual-reporter imaging can improve sensitivity and specificity over conventional fluorescence imaging in applications such as fluorescence-guided surgery and directly approximates the receptor status of the tumor, a measure that could be used to inform choices of biological therapies

    Quantitative Imaging of Scattering Changes Associated with Epithelial Proliferation, Necrosis and Fibrosis in Tumors Using Microsampling Reflectance Spectroscopy

    Get PDF
    Highly localized reflectance measurements can be used to directly quantify scatter changes in tissues. We present a microsampling approach that is used to raster scan tumors to extract parameters believed to be related to the tissue ultrastructure. A confocal reflectance imager was developed to examine scatter changes across pathologically distinct regions within tumor tissues. Tissue sections from two murine tumors, AsPC-1 pancreas tumor and the Mat-LyLu Dunning prostate tumor, were imaged. After imaging, histopathology-guided region-of-interest studies of the images allowed analysis of the variations in scattering resulting from differences in tissue ultra-structure. On average, the median scatter power of tumor cells with high proliferation index (HPI) was about 26% less compared to tumor cells with low proliferation index (LPI). Necrosis exhibited the lowest scatter power signature across all the tissue types considered, with about 55% lower median scatter power than LPI tumor cells. Additionally, the level and maturity of the tumor\u27s fibroplastic response was found to influence the scatter signal. This approach to scatter visualization of tissue ultrastructure in situ could provide a unique tool for guiding surgical resection, but this kind of interpretation into what the signal means relative to the pathology is required before proceeding to clinical studies

    Imaging Targeted-Agent Binding In Vivo with Two Probes

    Get PDF
    An approach to quantitatively image targeted-agent binding rate in vivo is demonstrated with dual-probe injection of both targeted and nontargeted fluorescent dyes. Images of a binding rate constant are created that reveal lower than expected uptake of epidermal growth factor in an orthotopic xenograft pancreas tumor (2.3×10−5 s−1), as compared to the normal pancreas (3.4×10−5 s−1). This approach allows noninvasive assessment of tumor receptor targeting in vivo to determine the expected contrast, spatial localization, and efficacy in therapeutic agent delivery. Targeting therapeutic drugs to tumors based on their overexpression of cellular receptors is widely researched and has important clinical success.1, 2 Yet there are essentially no good tools to assess the in vivo receptor expression contrast between tumor as compared to normal surrounding tissue.3, 4 In tumors with very high molecular signaling such as in the pancreas,4, 5 it is not obvious when a particular receptor is actually up-regulated as compared to the surrounding normal tissue versus upregulated without biopsy. Imaging of receptor status in vivo is problematic, because the majority of any targeted agent in vivo is often not cell-associated yet. Thus, any single image simply provides a measure of the whole tissue concentration rather than the bound concentration. Delivery from the vascular supply to tumor cells requires transvascular leakage, followed by diffusion through the interstitial space, and binding to the targeted receptor followed by possible internalization.6 As such, imaging concentration values in vivo usually do not provide information about binding,7 since most of the agent is in the interstitial space. In this work, we demonstrate a new methodology for quantitative imaging of effective binding rate in vivo, using the difference in fluorescence signal between a targeted and untargeted agent. We use this to demonstrate that a tumor known to have high EGFR expression in vitro 5 actually has lower EGF activity than the surrounding normal pancreas in vivo. Most contrast agent imaging has been interpreted with a simple pharmacokinetic model that is designed with as few compartments and rate constants as possible to not overinterpret the data. A three compartment model [Fig. 1 ] can be used effectively to model targeted agent delivery in the tumor, which includes compartments for 1. the concentration of drug in the plasma within the vasculature, 2. the concentration in the interstitial space of the tissue, and 3. the cellular-associated fraction of drug.7 The dominant fast rates in this model are transvascular delivery of contrast agent through rate constantK12 role= presentation \u3eK12 , and then cell-associating rate constant due to binding and uptake, K23 role= presentation \u3eK23 . The dominant clearance from the plasma is given by excretion mechanisms, such as those in the liver and kidneys, through rate constant Ke role= presentation \u3eKe . Then the slowest rates tend to be those involved in backflow from the interstitial space to the vasculature K21 role= presentation \u3eK21 , and from the cell-associated space to the interstitial space K32 role= presentation \u3eK32 . Each of these is shown in the illustration of the model in Fig. 1

    3D printing fluorescent material with tunable optical properties

    Get PDF
    The 3D printing of fluorescent materials could help develop, validate, and translate imaging technologies, including systems for fluorescence-guided surgery. Despite advances in 3D printing techniques for optical targets, no comprehensive method has been demonstrated for the simultaneous incorporation of fluorophores and fine-tuning of absorption and scattering properties. Here, we introduce a photopolymer-based 3D printing method for manufacturing fluorescent material with tunable optical properties. The results demonstrate the ability to 3D print various individual fluorophores at reasonably high fluorescence yields, including IR-125, quantum dots, methylene blue, and rhodamine 590. Furthermore, tuning of the absorption and reduced scattering coefficients is demonstrated within the relevant mamalian soft tissue coefficient ranges of 0.005–0.05 mm−1 and 0.2–1.5 mm−1, respectively. Fabrication of fluorophore-doped biomimicking and complex geometric structures validated the ability to print feature sizes less than 200 μm. The presented methods and optical characterization techniques provide the foundation for the manufacturing of solid 3D printed fluorescent structures, with direct relevance to biomedical optics and the broad adoption of fast manufacturing methods in fluorescence imaging

    Automated interpretation of scatter signatures aimed at tissue morphology identification

    Get PDF
    An automated algorithm and methodology is presented to pathologically classify the scattering changes encountered in the raster scanning of normal and tumor pancreatic tissues using microsampling reflectance spectroscopy. A quasiconfocal reflectance imaging system was used to directly measure the tissue scatter reflectance in situ, and the spectrum was used to identify the scattering power, amplitude and total wavelength-integrated intensity. Pancreatic tumor and normal samples were characterized using the instrument and subtle changes in the scatter signal were encountered within regions of each sample. Discrimination between normal vs. tumor tissue was readily performed using an Artificial Neural Network (ANN) classifier algorithm. A similar approach has worked also for regions of tumor morphology when statistical pre-processing of the scattering parameters was included to create additional data features. This automated interpretation methodology can provide a tool for guiding surgical resection in areas where microscopy imaging do not reach enough contrast to assist the surgeon
    • …
    corecore