Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Lab Invest ; 104(6): 102060, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38626875

ABSTRACT

Precision medicine aims to provide personalized care based on individual patient characteristics, rather than guideline-directed therapies for groups of diseases or patient demographics. Images-both radiology- and pathology-derived-are a major source of information on presence, type, and status of disease. Exploring the mathematical relationship of pixels in medical imaging ("radiomics") and cellular-scale structures in digital pathology slides ("pathomics") offers powerful tools for extracting both qualitative and, increasingly, quantitative data. These analytical approaches, however, may be significantly enhanced by applying additional methods arising from fields of mathematics such as differential geometry and algebraic topology that remain underexplored in this context. Geometry's strength lies in its ability to provide precise local measurements, such as curvature, that can be crucial for identifying abnormalities at multiple spatial levels. These measurements can augment the quantitative features extracted in conventional radiomics, leading to more nuanced diagnostics. By contrast, topology serves as a robust shape descriptor, capturing essential features such as connected components and holes. The field of topological data analysis was initially founded to explore the shape of data, with functional network connectivity in the brain being a prominent example. Increasingly, its tools are now being used to explore organizational patterns of physical structures in medical images and digitized pathology slides. By leveraging tools from both differential geometry and algebraic topology, researchers and clinicians may be able to obtain a more comprehensive, multi-layered understanding of medical images and contribute to precision medicine's armamentarium.


Subject(s)
Precision Medicine , Precision Medicine/methods , Humans , Radiology/methods , Image Processing, Computer-Assisted/methods
2.
Am J Pathol ; 192(2): 180-194, 2022 02.
Article in English | MEDLINE | ID: mdl-34774514

ABSTRACT

Conventional analysis using clinical histopathology is based on bright-field microscopy of thinly sliced tissue specimens. Although bright-field microscopy is a simple and robust method of examining microscope slides, the preparation of the slides needed is a lengthy and labor-intensive process. Slide-free histopathology, however, uses direct imaging of intact, minimally processed tissue samples using advanced optical-imaging systems, bypassing the extended workflow now required for the preparation of tissue sections. This article explains the technical basis of slide-free microscopy, reviews common slide-free optical microscopy techniques, and discusses the opportunities and challenges involved in clinical implementation.


Subject(s)
Image Processing, Computer-Assisted , Microscopy , Pathology, Clinical , Humans
3.
Transpl Int ; 36: 11783, 2023.
Article in English | MEDLINE | ID: mdl-37908675

ABSTRACT

The Banff Digital Pathology Working Group (DPWG) was established with the goal to establish a digital pathology repository; develop, validate, and share models for image analysis; and foster collaborations using regular videoconferencing. During the calls, a variety of artificial intelligence (AI)-based support systems for transplantation pathology were presented. Potential collaborations in a competition/trial on AI applied to kidney transplant specimens, including the DIAGGRAFT challenge (staining of biopsies at multiple institutions, pathologists' visual assessment, and development and validation of new and pre-existing Banff scoring algorithms), were also discussed. To determine the next steps, a survey was conducted, primarily focusing on the feasibility of establishing a digital pathology repository and identifying potential hosts. Sixteen of the 35 respondents (46%) had access to a server hosting a digital pathology repository, with 2 respondents that could serve as a potential host at no cost to the DPWG. The 16 digital pathology repositories collected specimens from various organs, with the largest constituent being kidney (n = 12,870 specimens). A DPWG pilot digital pathology repository was established, and there are plans for a competition/trial with the DIAGGRAFT project. Utilizing existing resources and previously established models, the Banff DPWG is establishing new resources for the Banff community.


Subject(s)
Artificial Intelligence , Kidney Transplantation , Humans , Algorithms , Kidney/pathology
4.
Vet Pathol ; 60(1): 52-59, 2023 01.
Article in English | MEDLINE | ID: mdl-36286074

ABSTRACT

Fluorescence imitating brightfield imaging (FIBI) is a novel microscopy method that allows for real-time, nondestructive, slide-free tissue imaging of fresh, formalin-fixed, or paraffin-embedded tissue. The nondestructive nature of the technology permits tissue preservation for downstream analyses. The objective of this observational study was to assess the utility of FIBI compared with conventional hematoxylin and eosin (H&E)-stained histology slides in feline gastrointestinal histopathology. Formalin-fixed paraffin-embedded full-thickness small intestinal tissue specimens from 50 cases of feline chronic enteropathy were evaluated. The ability of FIBI to evaluate predetermined morphological features (epithelium, villi, crypts, lacteals, fibrosis, submucosa, and muscularis propria) and inflammatory cells was assessed on a 3-point scale (0 = FIBI cannot identify the feature; 1 = FIBI can identify the feature; 2 = FIBI can identify the feature with more certainty than H&E). H&E and FIBI images were also scored according to World Small Animal Veterinary Association (WSAVA) Gastrointestinal Standardization Group guidelines. FIBI identified morphological features with similar or, in some cases, higher confidence compared with H&E images. The identification of inflammatory cells was less consistent. FIBI and H&E images showed an overall poor agreement with regard to the assigned WSAVA scores. While FIBI showed an equal or better ability to identify morphological features in intestinal biopsies, its ability to identify inflammatory cells is currently inferior compared with H&E-based imaging. Future studies on the utility of FIBI as a diagnostic tool for noninflammatory histopathologic lesions are warranted.


Subject(s)
Cat Diseases , Inflammatory Bowel Diseases , Cats , Animals , Microscopy/veterinary , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/veterinary , Intestine, Small/pathology , Duodenum/pathology , Biopsy/veterinary , Cat Diseases/diagnostic imaging , Cat Diseases/pathology
5.
Mod Pathol ; 35(10): 1362-1369, 2022 10.
Article in English | MEDLINE | ID: mdl-35729220

ABSTRACT

Ki67 has potential clinical importance in breast cancer but has yet to see broad acceptance due to inter-laboratory variability. Here we tested an open source and calibrated automated digital image analysis (DIA) platform to: (i) investigate the comparability of Ki67 measurement across corresponding core biopsy and resection specimen cases, and (ii) assess section to section differences in Ki67 scoring. Two sets of 60 previously stained slides containing 30 core-cut biopsy and 30 corresponding resection specimens from 30 estrogen receptor-positive breast cancer patients were sent to 17 participating labs for automated assessment of average Ki67 expression. The blocks were centrally cut and immunohistochemically (IHC) stained for Ki67 (MIB-1 antibody). The QuPath platform was used to evaluate tumoral Ki67 expression. Calibration of the DIA method was performed as in published studies. A guideline for building an automated Ki67 scoring algorithm was sent to participating labs. Very high correlation and no systematic error (p = 0.08) was found between consecutive Ki67 IHC sections. Ki67 scores were higher for core biopsy slides compared to paired whole sections from resections (p ≤ 0.001; median difference: 5.31%). The systematic discrepancy between core biopsy and corresponding whole sections was likely due to pre-analytical factors (tissue handling, fixation). Therefore, Ki67 IHC should be tested on core biopsy samples to best reflect the biological status of the tumor.


Subject(s)
Breast Neoplasms , Biomarkers, Tumor/analysis , Biopsy , Breast Neoplasms/pathology , Female , Humans , Image Processing, Computer-Assisted/methods , Immunohistochemistry , Ki-67 Antigen/analysis , Receptors, Estrogen
6.
Am J Transplant ; 20(9): 2392-2399, 2020 09.
Article in English | MEDLINE | ID: mdl-32185875

ABSTRACT

The Banff Digital Pathology Working Group (DPWG) was formed in the time leading up to and during the joint American Society for Histocompatibility and Immunogenetics/Banff Meeting, September 23-27, 2019, held in Pittsburgh, Pennsylvania. At the meeting, the 14th Banff Conference, presentations directly and peripherally related to the topic of "digital pathology" were presented; and discussions before, during, and after the meeting have resulted in a list of issues to address for the DPWG. Included are practice standardization, integrative approaches for study classification, scoring of histologic parameters (eg, interstitial fibrosis and tubular atrophy and inflammation), algorithm classification, and precision diagnosis (eg, molecular pathways and therapeutics). Since the meeting, a survey with international participation of mostly pathologists (81%) was conducted, showing that whole slide imaging is available at the majority of centers (71%) but that artificial intelligence (AI)/machine learning was only used in ≈12% of centers, with a wide variety of programs/algorithms employed. Digitalization is not just an end in itself. It also is a necessary precondition for AI and other approaches. Discussions at the meeting and the survey highlight the unmet need for a Banff DPWG and point the way toward future contributions that can be made.


Subject(s)
Kidney Diseases , Kidney Transplantation , Artificial Intelligence , Biopsy , Graft Rejection , Humans , Pennsylvania
7.
J Drugs Dermatol ; 15(11): 1344-1346, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-28095545

ABSTRACT

BACKGROUND: Skin care products make up the largest part (36%) of the cosmetic market globally, of which the United States plays the largest role. In 2015, approximately 115 billion USD was spent globally on skin care products. Skin care products, in contradistinction to pharmaceuticals, are not strictly regulated by the FDA. A key factor for evaluation of a skin care product or topical drug is skin barrier function and effect on super cial skin. Thus, it is critical to have quantitative and qualitative methods to study the effects of skin care products on skin barrier and the super cial skin. Currently, no imaging method exists that can evaluate and track super cial skin changes visually in real-time. OBJECTIVE: To report using a novel imaging modality, Microscopy using Ultraviolet Surface Excitation (MUSE), to provide real-time, high- resolution, in vivo characterization of super cial skin and moisturizing properties of topical moisturizer, and to highlight key bene ts of using MUSE to visualize the super cial skin and serve as an excellent complementary tool to current quantitative methods. METHODS AND MATERIALS: The methodology of MUSE is based upon two main principles inherent to ultraviolet (UV) light and uorescent staining agents. In this study, the author's (JJ) index ngertip was imaged using the MUSE instrument without and with moisturizer. RESULTS: Dermatoglyphics of the fingertip consists of ridges (cristae super ciales) and grooves (sulci super ciales) proved to be straightforward to visualize at high resolution. Desquamation of superficial corneocytes and opening of an acrosyringium (the most superficial portion of eccrine ducts) were visualized in high-resolution. Post-application of a moisturizer, a uniform layer of moisturizer could be seen superficial to the corneocytes along the ridges and CONCLUSIONS: Real-time, high-resolution, in vivo characterization of super cial skin and moisturizing properties of moisturizer using MUSE is feasible. Its utility can be enhanced with downstream quantification using imaging software. J Drugs Dermatol. 2016;15(11):1344-1346..


Subject(s)
Epidermal Cells , Epidermis/drug effects , Microscopy , Skin Cream/administration & dosage , Ultraviolet Rays , Cosmetics/administration & dosage , Humans , Skin/cytology , Skin/drug effects
8.
Lab Invest ; 95(4): 397-405, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25730370

ABSTRACT

The role of immunohistochemistry (IHC) in the management of cancer has expanded to provide improved diagnostic classification, as well as guidance on disease prognosis, therapy, and relapse. These new tasks require evaluation of an increasing number of protein targets; however, conventional multiplexing, usually achieved using serial tissue sections stained for a single analyte per slide, can exhaust small biopsy specimens, complicate slide-to-slide protein expression correlation, and leave insufficient material for additional molecular assays. A new approach, mass spectrometry immunohistochemistry (MSIHC), compatible with high levels of target multiplexing and suitable for use on formalin-fixed, paraffin-embedded samples can circumvent many of these issues. The strategy employs antibodies that are labeled with elemental mass tags, such as isotopically pure lanthanides not typically found in biological specimens, rather than with typical fluorophores or chromogens. The metal-labeled antibodies are then detected in tissue using lasers or ion beams to liberate the tags for subsequent mass spectrometry detection. Within a given multiplexed IHC panel, the metal labels are selected so that their respective masses do not overlap. More than 30 antibodies have been imaged simultaneously, and up to 100 antibodies could potentially be detected at once if the full available mass spectrum is deployed. MSIHC has a number of advantages over conventional IHC techniques. Background due to autofluorescence is absent and the dynamic range is 10(5), exceeding immunofluorescence and chromogenic IHC by 100-fold and 1000-fold, respectively. Detection of labeled primary antibodies improves assay linearity over both chromogenic and fluorescent IHC. Multiplexed mass-tagged antibodies incubated simultaneously with tissue do not appear to cross-interfere, and because the mass tags do not degrade, samples are stable indefinitely. The imaging resolution of multiplexed ion-beam imaging can be better than light microscopy. With appropriate instrumentation, MSIHC has the potential to transform research and clinical pathology practice.


Subject(s)
Immunohistochemistry/methods , Mass Spectrometry/methods , Molecular Imaging/methods , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/chemistry , Humans , Mice , Neoplasms/chemistry , Neoplasms/diagnosis , Neoplasms/pathology
9.
Arch Pathol Lab Med ; 148(3): 345-352, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37226827

ABSTRACT

CONTEXT.­: Digital pathology using whole slide images has been recently approved to support primary diagnosis in clinical surgical pathology practices. Here we describe a novel imaging method, fluorescence-imitating brightfield imaging, that can capture the surface of fresh tissue without requiring prior fixation, paraffin embedding, tissue sectioning, or staining. OBJECTIVE.­: To compare the ability of pathologists to evaluate direct-to-digital images with standard pathology preparations. DESIGN.­: One hundred surgical pathology samples were obtained. Samples were first digitally imaged, then processed for standard histologic examination on 4-µm hematoxylin-eosin-stained sections and digitally scanned. The resulting digital images from both digital and standard scan sets were viewed by each of 4 reading pathologists. The data set consisted of 100 reference diagnoses and 800 study pathologist reads. Each study read was compared to the reference diagnosis, and also compared to that reader's diagnosis across both modalities. RESULTS.­: The overall agreement rate, across 800 reads, was 97.9%. This consisted of 400 digital reads at 97.0% versus reference and 400 standard reads versus reference at 98.8%. Minor discordances (defined as alternative diagnoses without clinical treatment or outcome implications) were 6.1% overall, 7.2% for digital, and 5.0% for standard. CONCLUSIONS.­: Pathologists can provide accurate diagnoses from fluorescence-imitating brightfield imaging slide-free images. Concordance and discordance rates are similar to published rates for comparisons of whole slide imaging to standard light microscopy of glass slides for primary diagnosis. It may be possible, therefore, to develop a slide-free, nondestructive approach for primary pathology diagnosis.


Subject(s)
Pathology, Surgical , Humans , Hematoxylin , Eosine Yellowish-(YS) , Pathology, Surgical/methods , Paraffin Embedding , Microscopy/methods , Formaldehyde
10.
PLoS One ; 18(8): e0289139, 2023.
Article in English | MEDLINE | ID: mdl-37552656

ABSTRACT

The rapid emergence and global dissemination of SARS-CoV-2 that causes COVID-19 continues to cause an unprecedented global health burden resulting in nearly 7 million deaths. While multiple vaccine countermeasures have been approved for emergency use, additional treatments are still needed due to sluggish vaccine rollout, vaccine hesitancy, and inefficient vaccine-mediated protection. Immunoadjuvant compounds delivered intranasally can guide non-specific innate immune responses during the critical early stages of viral replication, reducing morbidity and mortality. N-dihydrogalactochitosan (GC) is a novel mucoadhesive immunostimulatory polymer of ß-0-4-linked N-acetylglucosamine that is solubilized by the conjugation of galactose glycans with current applications as a cancer immunotherapeutic. We tested GC as a potential countermeasure for COVID-19. GC was well-tolerated and did not produce histopathologic lesions in the mouse lung. GC administered intranasally before and after SARS-CoV-2 exposure diminished morbidity and mortality in humanized ACE2 receptor expressing mice by up to 75% and reduced infectious virus levels in the upper airway. Fluorescent labeling of GC shows that it is confined to the lumen or superficial mucosa of the nasal cavity, without involvement of adjacent or deeper tissues. Our findings demonstrate a new application for soluble immunoadjuvants such as GC for preventing disease associated with SARS-CoV-2 and may be particularly attractive to persons who are needle-averse.


Subject(s)
COVID-19 , SARS-CoV-2 , Mice , Animals , Acetylglucosamine , Virus Replication
11.
Sci Rep ; 12(1): 10205, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35715554

ABSTRACT

Understanding peripheral nerve micro-anatomy can assist in the development of safe and effective neuromodulation devices. However, current approaches for imaging nerve morphology at the fiber level are either cumbersome, require substantial instrumentation, have a limited volume of view, or are limited in resolution/contrast. We present alternative methods based on MUSE (Microscopy with Ultraviolet Surface Excitation) imaging to investigate peripheral nerve morphology, both in 2D and 3D. For 2D imaging, fixed samples are imaged on a conventional MUSE system either label free (via auto-fluorescence) or after staining with fluorescent dyes. This method provides a simple and rapid technique to visualize myelinated nerve fibers at specific locations along the length of the nerve and perform measurements of fiber morphology (e.g., axon diameter and g-ratio). For 3D imaging, a whole-mount staining and MUSE block-face imaging method is developed that can be used to characterize peripheral nerve micro-anatomy and improve the accuracy of computational models in neuromodulation. Images of rat sciatic and human cadaver tibial nerves are presented, illustrating the applicability of the method in different preclinical models.


Subject(s)
Alprostadil , Peripheral Nerves , Animals , Axons , Imaging, Three-Dimensional/methods , Nerve Fibers, Myelinated , Peripheral Nerves/diagnostic imaging , Rats , Sciatic Nerve/diagnostic imaging
12.
Lab Chip ; 22(7): 1354-1364, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35212692

ABSTRACT

Minimally invasive core needle biopsies for medical diagnoses have become increasingly common for many diseases. Although tissue cores can yield more diagnostic information than fine needle biopsies and cytologic evaluations, there is no rapid assessment at the point-of-care for intact tissue cores that is low-cost and non-destructive to the biopsy. We have developed a proof-of-concept 3D printed millifluidic histopathology lab-on-a-chip device to automatically handle, process, and image fresh core needle biopsies. This device, named CoreView, includes modules for biopsy removal from the acquisition tool, transport, staining and rinsing, imaging, segmentation, and multiplexed storage. Reliable removal from side-cutting needles and bidirectional fluid transport of core needle biopsies of five tissue types has been demonstrated with 0.5 mm positioning accuracy. Automation is aided by a MATLAB-based biopsy tracking algorithm that can detect the location of tissue and air bubbles in the channels of the millifluidic chip. With current and emerging optical imaging technologies, CoreView can be used for a rapid adequacy test at the point-of-care for tissue identification as well as glomeruli counting in renal core needle biopsies.


Subject(s)
Algorithms , Kidney , Biopsy , Biopsy, Large-Core Needle
13.
Commun Biol ; 4(1): 334, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712728

ABSTRACT

Smartphone microscopes can be useful tools for a broad range of imaging applications. This manuscript demonstrates the first practical implementation of Microscopy with Ultraviolet Surface Excitation (MUSE) in a compact smartphone microscope called Pocket MUSE, resulting in a remarkably effective design. Fabricated with parts from consumer electronics that are readily available at low cost, the small optical module attaches directly over the rear lens in a smartphone. It enables high-quality multichannel fluorescence microscopy with submicron resolution over a 10× equivalent field of view. In addition to the novel optical configuration, Pocket MUSE is compatible with a series of simple, portable, and user-friendly sample preparation strategies that can be directly implemented for various microscopy applications for point-of-care diagnostics, at-home health monitoring, plant biology, STEM education, environmental studies, etc.


Subject(s)
Microscopy, Fluorescence/instrumentation , Mobile Applications , Smartphone , Animals , Bacteria , Equipment Design , Fluorescent Antibody Technique , Humans , Image Processing, Computer-Assisted , Mice , Mouth Mucosa , Plants , Reproducibility of Results , Specimen Handling , Staining and Labeling
14.
Sci Rep ; 11(1): 19063, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34561546

ABSTRACT

Over the past two decades, fibrillar collagen reorganization parameters such as the amount of collagen deposition, fiber angle and alignment have been widely explored in numerous studies. These parameters are now widely accepted as stromal biomarkers and linked to disease progression and survival time in several cancer types. Despite all these advances, there has not been a significant effort to make it possible for clinicians to explore these biomarkers without adding steps to the clinical workflow or by requiring high-cost imaging systems. In this paper, we evaluate previously described polychromatic polarization microscope (PPM) to visualize collagen fibers with an optically generated color representation of fiber orientation and alignment when inspecting the sample by a regular microscope with minor modifications. This system does not require stained slides, but is compatible with histological stains such as H&E. Consequently, it can be easily accommodated as part of regular pathology review of tissue slides, while providing clinically useful insight into stromal composition.


Subject(s)
Fibrillar Collagens/metabolism , Microscopy, Polarization/methods , Adenocarcinoma/metabolism , Biomarkers/metabolism , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Female , Humans , Male , Pancreas/metabolism , Pancreas/pathology , Prostatic Neoplasms/metabolism
15.
Bioconjug Chem ; 20(7): 1367-74, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19514716

ABSTRACT

In vivo fluorescence cancer imaging is an important tool in understanding tumor growth and therapeutic monitoring and can be performed either with endogenously produced fluorescent proteins or with exogenously introduced fluorescent probes bound to targeting molecules. However, endogenous fluorescence proteins cannot be altered after transfection, thus requiring rederivation of cell lines for each desired color, while exogenously targeted fluorescence probes are limited by the heterogeneous expression of naturally occurring cellular targets. In this study, we adapted the dehalogenase-based protein-Tag (HaloTag) system to in vivo cancer imaging, by introducing highly expressed HaloTag receptors (HaloTagR) in cancer cells coupled with a range of externally injected fluorophore-conjugated dehalogenase-reactive reactive linkers. Tumor nodules arising from a single transfected cell line were stably labeled with fluorescence varying in emission spectra from green to near-infrared. After establishing and validating a SHIN3 cell line stably transfected with HaloTagR (HaloTagR-SHIN3), in vivo spectral fluorescence imaging studies were performed in live animals using a peritoneal dissemination model. The tumor nodules arising from HaloTagR-SHIN3 could be successfully labeled by four different fluorophore-conjugated HaloTag-ligands each emitting light at different wavelengths. These fluorophores could be alternated on serial imaging sessions permitting assessment of interval growth. Fluorescence was retained in histological specimens after fixation. Thus, this tagging system proves versatile both for in vivo and in vitro imaging without requiring modification of the underlying cell line. Thus, this strategy can overcome some of the limitations associated with the use of endogenous fluorescent proteins and exogenous targeted optical agents in current use.


Subject(s)
Diagnostic Imaging/methods , Fluorescent Dyes/analysis , Ovarian Neoplasms/diagnosis , Proteins/analysis , Proteins/genetics , Animals , Binding Sites , Cell Line, Tumor , Endoscopy , Female , Fluorescence , Gene Expression , Humans , Ligands , Mice , Ovarian Neoplasms/pathology , Transfection
16.
Arch Pathol Lab Med ; 143(9): 1058-1068, 2019 09.
Article in English | MEDLINE | ID: mdl-31295016

ABSTRACT

CONTEXT.­: The rapid evolution of optical imaging modalities in recent years has opened the opportunity for ex vivo tissue imaging, which has significant implications for surgical pathology practice. These modalities have promising potential to be used as next-generation digital microscopy tools for examination of fresh tissue, with or without labeling with contrast agents. OBJECTIVE.­: To review the literature regarding various types of ex vivo optical imaging platforms that can generate digital images for tissue recognition with potential for utilization in anatomic pathology clinical practices. DATA SOURCES.­: Literature relevant to ex vivo tissue imaging obtained from the PubMed database. CONCLUSIONS.­: Ex vivo imaging of tissues can be performed by using various types of optical imaging techniques. These next-generation digital microscopy tools have a promising potential for utilization in surgical pathology practice.


Subject(s)
Microscopy , Optical Imaging , Pathology, Surgical , Humans , Clinical Laboratory Techniques , Microscopy/instrumentation , Microscopy/methods , Microscopy/trends , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Ultraviolet , Nonlinear Optical Microscopy , Optical Imaging/instrumentation , Optical Imaging/methods , Optical Imaging/trends , Pathology, Clinical/methods , Pathology, Surgical/instrumentation , Pathology, Surgical/methods , Pathology, Surgical/trends , Tomography, Optical Coherence
17.
ILAR J ; 49(1): 78-88, 2008.
Article in English | MEDLINE | ID: mdl-18172335

ABSTRACT

Increasing sophistication in the design and application of biological models as well as the advent of novel fluorescent probes have led to new demands on molecular imaging systems to deliver enhanced sensitivity, reliable quantitation, and the ability to resolve multiple simultaneous signals. Sensitivity is limited, especially in the visible spectral range, by the presence of ubiquitous autofluorescence signals (mostly arising from the skin and gut), which need to be separated from those of targeted fluorophores. Fluorescence-based imaging is also affected by absorbing and scattering properties of tissue in both the visible and to a lesser extent the near-infrared (NIR) regions. However, the small size of typical animal models (usually mice) often permits the detection of enough light arising even from relatively deep locations to allow the capture of signals with an acceptable signal-to-noise ratio. Multispectral imaging, through its ability to separate autofluorescence from label fluorescence, can increase sensitivity as much as 300 times compared to conventional approaches, and concomitantly improve quantitative accuracy. In the NIR region, autofluorescence, while still significant, poses less of a problem. However, the task of disentangling signals from multiple fluorophores remains. Multispectral imaging allows the separation of five or more fluorophores, with each signal quantitated and visualized separately. Preclinical small animal imaging is often accompanied by microscopic analysis, both before and after the in vivo phase. This can involve tissue culture manipulations and/or histological examination of fixed or frozen tissue. Due to the same advantages in sensitivity, quantitation, and multiplexing, microscopy-based multispectral techniques form an excellent complement to in vivo imaging.


Subject(s)
Fluorescence , Microscopy, Fluorescence/methods , Animals , Fluorescent Dyes/chemistry , Image Enhancement/methods , Mice , Reproducibility of Results
18.
J Mol Diagn ; 9(1): 20-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17251332

ABSTRACT

Gene expression profiling has identified several potentially useful gene signatures for predicting outcome or for selecting targeted therapy. However, these signatures have been developed in fresh or frozen tissue, and there is a need to apply them to routinely processed samples. Here, we demonstrate the feasibility of a potentially high-throughput methodology combining automated in situ hybridization with quantum dot-labeled oligonucleotide probes followed by spectral imaging for the detection and subsequent deconvolution of multiple signals. This method is semiautomated and quantitative and can be applied to formalin-fixed, paraffin-embedded tissues. We have combined dual in situ hybridization with immunohistochemistry, enabling simultaneous measurement of gene expression and cell lineage determination. The technique achieves levels of sensitivity and specificity sufficient for the potential application of known expression signatures to biopsy specimens in a semiquantitative way, and the semiautomated nature of the method enables application to high-throughput studies.


Subject(s)
Cell Lineage , Gene Expression Profiling/methods , In Situ Hybridization/methods , Molecular Diagnostic Techniques/methods , Quantum Dots , Animals , DNA, Complementary/genetics , Humans , Image Processing, Computer-Assisted , Immunohistochemistry/methods , Mice , Oligonucleotide Probes , Sensitivity and Specificity , Tumor Cells, Cultured
19.
J Biomed Opt ; 12(5): 051801, 2007.
Article in English | MEDLINE | ID: mdl-17994879

ABSTRACT

Noninvasive optical imaging technology has the potential to improve the accuracy of disease detection and predict treatment response. Pathology provides the critical link between the biological basis of an image or spectral signature and clinical outcomes obtained through optical imaging. The validation of optical images and spectra requires both morphologic diagnosis from histopathology and parametric analysis of tissue features above and beyond the declared pathologic "diagnosis." Enhancement of optical imaging modalities with exogenously applied biomarkers also requires validation of the biological basis for molecular contrast. For an optical diagnostic or prognostic technology to be useful, it must be clinically important, independently informative, and of demonstrated beneficial value to patient care. Its usage must be standardized with regard to methods, interpretation, reproducibility, and reporting, in which the pathologist plays a key role. By providing insight into disease pathobiology, interpretive or quantitative analysis of tissue material, and expertise in molecular diagnosis, the pathologist should be an integral part of any team that is validating novel optical imaging modalities. This review will consider (1) the selection of validation biomarkers; (2) standardization in tissue processing, diagnosis, reporting, and quantitative analysis; (3) the role of the pathologist in study design; and (4) reference standards, controls, and interobserver variability.


Subject(s)
Biotechnology/standards , Diagnostic Imaging/standards , Image Interpretation, Computer-Assisted/standards , Optics and Photonics , Pathology/standards , Practice Guidelines as Topic , Quality Assurance, Health Care/standards , United States
20.
Nat Biotechnol ; 22(8): 969-76, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258594

ABSTRACT

We describe the development of multifunctional nanoparticle probes based on semiconductor quantum dots (QDs) for cancer targeting and imaging in living animals. The structural design involves encapsulating luminescent QDs with an ABC triblock copolymer and linking this amphiphilic polymer to tumor-targeting ligands and drug-delivery functionalities. In vivo targeting studies of human prostate cancer growing in nude mice indicate that the QD probes accumulate at tumors both by the enhanced permeability and retention of tumor sites and by antibody binding to cancer-specific cell surface biomarkers. Using both subcutaneous injection of QD-tagged cancer cells and systemic injection of multifunctional QD probes, we have achieved sensitive and multicolor fluorescence imaging of cancer cells under in vivo conditions. We have also integrated a whole-body macro-illumination system with wavelength-resolved spectral imaging for efficient background removal and precise delineation of weak spectral signatures. These results raise new possibilities for ultrasensitive and multiplexed imaging of molecular targets in vivo.


Subject(s)
Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Drug Delivery Systems/methods , Microscopy, Fluorescence/methods , Neoplasms/metabolism , Neoplasms/pathology , Quantum Dots , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Image Enhancement/methods , Male , Materials Testing , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Semiconductors , Spectrometry, Fluorescence/methods , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL