Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Front Mol Biosci ; 10: 1051491, 2023.
Article in English | MEDLINE | ID: mdl-36845550

ABSTRACT

Immunohistochemistry has long been held as the gold standard for understanding the expression patterns of therapeutically relevant proteins to identify prognostic and predictive biomarkers. Patient selection for targeted therapy in oncology has successfully relied upon standard microscopy-based methodologies, such as single-marker brightfield chromogenic immunohistochemistry. As promising as these results are, the analysis of one protein, with few exceptions, no longer provides enough information to draw effective conclusions about the probability of treatment response. More multifaceted scientific queries have driven the development of high-throughput and high-order technologies to interrogate biomarker expression patterns and spatial interactions between cell phenotypes in the tumor microenvironment. Such multi-parameter data analysis has been historically reserved for technologies that lack the spatial context that is provided by immunohistochemistry. Over the past decade, technical developments in multiplex fluorescence immunohistochemistry and discoveries made with improving image data analysis platforms have highlighted the importance of spatial relationships between certain biomarkers in understanding a patient's likelihood to respond to, typically, immune checkpoint inhibitors. At the same time, personalized medicine has instigated changes in both clinical trial design and its conduct in a push to make drug development and cancer treatment more efficient, precise, and economical. Precision medicine in immuno-oncology is being steered by data-driven approaches to gain insight into the tumor and its dynamic interaction with the immune system. This is particularly necessary given the rapid growth in the number of trials involving more than one immune checkpoint drug, and/or using those in combination with conventional cancer treatments. As multiplex methods, like immunofluorescence, push the boundaries of immunohistochemistry, it becomes critical to understand the foundation of this technology and how it can be deployed for use as a regulated test to identify the prospect of response from mono- and combination therapies. To that end, this work will focus on: 1) the scientific, clinical, and economic requirements for developing clinical multiplex immunofluorescence assays; 2) the attributes of the Akoya Phenoptics workflow to support predictive tests, including design principles, verification, and validation needs; 3) regulatory, safety and quality considerations; 4) application of multiplex immunohistochemistry through lab-developed-tests and regulated in vitro diagnostic devices.

2.
Front Mol Biosci ; 8: 674747, 2021.
Article in English | MEDLINE | ID: mdl-34150850

ABSTRACT

As immuno-oncology (I/O) emerges as an effective approach in the fight against cancer, multispectral imaging of multiplex immunofluorescence (mIF) is maturing as an analytical platform. The timing is fortuitous. Due to health economic considerations surrounding the use of I/O, there is an urgent need for tests that accurately predict response to the growing list of available therapies. Multispectral mIF provides several advantages over other biomarker modalities by enabling deeper interrogation of the intricate biology within the tumor microenvironment, including detection of cell-to-cell spatial interactions that correlate with clinical outcomes. It also provides a practical path for generating reliable and reproducible results in a clinically suitable, high-throughput workflow. In this article, we (1) describe the principles behind multispectral mIF; (2) provide advice and recommendations on assay development and optimization and highlight characteristics of a well-performing assay; and (3) discuss the requirements for translating this approach into clinical practice.

3.
JACC Basic Transl Sci ; 5(4): 328-340, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32368693

ABSTRACT

Recognizing that guideline-directed histologic grading of endomyocardial biopsy tissue samples for rejection surveillance has limited diagnostic accuracy, quantitative, in situ characterization was performed of several important immune cell types in a retrospective cohort of clinical endomyocardial tissue samples. Differences between cases were identified and were grouped by histologic grade versus clinical rejection trajectory, with significantly increased programmed death ligand 1+, forkhead box P3+, and cluster of differentiation 68+ cells suppressed in clinically evident rejections, especially cases with marked clinical-histologic discordance. Programmed death ligand 1+, forkhead box P3+, and cluster of differentiation 68+ cell proportions are also significantly higher in "never-rejection" when compared with "future-rejection." These findings suggest that in situ immune modulators regulate the severity of cardiac allograft rejection.

4.
J Mammary Gland Biol Neoplasia ; 25(4): 417-432, 2020 12.
Article in English | MEDLINE | ID: mdl-33590360

ABSTRACT

Multiplex immunofluorescence (mIF) allows simultaneous antibody-based detection of multiple markers with a nuclear counterstain on a single tissue section. Recent studies have demonstrated that mIF is becoming an important tool for immune profiling the tumor microenvironment, further advancing our understanding of the interplay between cancer and the immune system, and identifying predictive biomarkers of response to immunotherapy. Expediting mIF discoveries is leading to improved diagnostic panels, whereas it is important that mIF protocols be standardized to facilitate their transition into clinical use. Manual processing of sections for mIF is time consuming and a potential source of variability across numerous samples. To increase reproducibility and throughput we demonstrate the use of an automated slide stainer for mIF incorporating tyramide signal amplification (TSA). We describe two panels aimed at characterizing the tumor immune microenvironment. Panel 1 included CD3, CD20, CD117, FOXP3, Ki67, pancytokeratins (CK), and DAPI, and Panel 2 included CD3, CD8, CD68, PD-1, PD-L1, CK, and DAPI. Primary antibodies were first tested by standard immunohistochemistry and single-plex IF, then multiplex panels were developed and images were obtained using a Vectra 3.0 multispectral imaging system. Various methods for image analysis (identifying cell types, determining cell densities, characterizing cell-cell associations) are outlined. These mIF protocols will be invaluable tools for immune profiling the tumor microenvironment.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/immunology , Fluoroimmunoassay/methods , Image Processing, Computer-Assisted/methods , Tumor Microenvironment/immunology , Biomarkers, Tumor/metabolism , Breast/immunology , Breast/pathology , Breast Neoplasms/pathology , Female , Fluorescent Dyes/chemistry , Fluoroimmunoassay/instrumentation , Humans , Reproducibility of Results , Tissue Array Analysis/instrumentation , Tissue Array Analysis/methods
5.
JAMA Oncol ; 5(8): 1195-1204, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31318407

ABSTRACT

IMPORTANCE: PD-L1 (programmed cell death ligand 1) immunohistochemistry (IHC), tumor mutational burden (TMB), gene expression profiling (GEP), and multiplex immunohistochemistry/immunofluorescence (mIHC/IF) assays have been used to assess pretreatment tumor tissue to predict response to anti-PD-1/PD-L1 therapies. However, the relative diagnostic performance of these modalities has yet to be established. OBJECTIVE: To compare studies that assessed the diagnostic accuracy of PD-L1 IHC, TMB, GEP, and mIHC/IF in predicting response to anti-PD-1/PD-L1 therapy. EVIDENCE REVIEW: A search of PubMed (from inception to June 2018) and 2013 to 2018 annual meeting abstracts from the American Association for Cancer Research, American Society of Clinical Oncology, European Society for Medical Oncology, and Society for Immunotherapy of Cancer was conducted to identify studies that examined the use of PD-L1 IHC, TMB, GEP, and mIHC/IF assays to determine objective response to anti-PD-1/PD-L1 therapy. For PD-L1 IHC, only clinical trials that resulted in US Food and Drug Administration approval of indications for anti-PD-1/PD-L1 were included. Studies combining more than 1 modality were also included. Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines were followed. Two reviewers independently extracted the clinical outcomes and test results for each individual study. MAIN OUTCOMES AND MEASURES: Summary receiver operating characteristic (sROC) curves; their associated area under the curve (AUC); and pooled sensitivity, specificity, positive and negative predictive values (PPV, NPV), and positive and negative likelihood ratios (LR+ and LR-) for each assay modality. RESULTS: Tumor specimens representing over 10 different solid tumor types in 8135 patients were assayed, and the results were correlated with anti-PD-1/PD-L1 response. When each modality was evaluated with sROC curves, mIHC/IF had a significantly higher AUC (0.79) compared with PD-L1 IHC (AUC, 0.65, P < .001), GEP (AUC, 0.65, P = .003), and TMB (AUC, 0.69, P = .049). When multiple different modalities were combined such as PD-L1 IHC and/or GEP + TMB, the AUC drew nearer to that of mIHC/IF (0.74). All modalities demonstrated comparable NPV and LR-, whereas mIHC/IF demonstrated higher PPV (0.63) and LR+ (2.86) than the other approaches. CONCLUSIONS AND RELEVANCE: In this meta-analysis, tumor mutational burden, PD-L1 IHC, and GEP demonstrated comparable AUCs in predicting response to anti-PD-1/PD-L1 treatment. Multiplex immunohistochemistry/IF and multimodality biomarker strategies appear to be associated with improved performance over PD-L1 IHC, TMB, or GEP alone. Further studies with mIHC/IF and composite approaches with a larger number of patients will be required to confirm these findings. Additional study is also required to determine the most predictive analyte combinations and to determine whether biomarker modality performance varies by tumor type.

6.
J Vis Exp ; (143)2019 01 21.
Article in English | MEDLINE | ID: mdl-30735177

ABSTRACT

Continued developments in immuno-oncology require an increased understanding of the mechanisms of cancer immunology. The immunoprofiling analysis of tissue samples from formalin-fixed, paraffin-embedded (FFPE) biopsies has become a key tool for understanding the complexity of tumor immunology and discovering novel predictive biomarkers for cancer immunotherapy. Immunoprofiling analysis of tissues requires the evaluation of combined markers, including inflammatory cell subpopulations and immune checkpoints, in the tumor microenvironment. The advent of novel multiplex immunohistochemical methods allows for a more efficient multiparametric analysis of single tissue sections than does standard monoplex immunohistochemistry (IHC). One commercially available multiplex immunofluorescence (IF) method is based on tyramide-signal amplification and, combined with multispectral microscopic analysis, allows for a better signal separation of diverse markers in tissue. This methodology is compatible with the use of unconjugated primary antibodies that have been optimized for standard IHC on FFPE tissue samples. Herein we describe in detail an automated protocol that allows multiplex IF labeling of carcinoma tissue samples with a six-marker multiplex antibody panel comprising PD-L1, PD-1, CD68, CD8, Ki-67, and AE1/AE3 cytokeratins with 4',6-diamidino-2-phenylindole as a nuclear cell counterstain. The multiplex panel protocol is optimized in an automated IHC stainer for a staining time that is shorter than that of the manual protocol and can be directly applied and adapted by any laboratory investigator for immuno-oncology studies on human FFPE tissue samples. Also described are several controls and tools, including a drop-control method for fine quality control of a new multiplex IF panel, that are useful for the optimization and validation of the technique.


Subject(s)
Carcinoma/pathology , Fluorescent Antibody Technique/methods , Formaldehyde/therapeutic use , Immunohistochemistry/methods , Humans , Tumor Microenvironment
7.
JCI Insight ; 2(14)2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28724788

ABSTRACT

Evaluation of T lymphocyte frequency provides prognostic information for patients with oral squamous cell cancer (OSCC). However, the effect of simultaneously evaluating T cell frequency and assessing suppressive elements and defects in antigen-processing machinery (APM) has not been clarified. Simultaneous characterization of CD3+, CD8+, FoxP3+, CD163+, and PD-L1+ cells using multispectral imaging was performed on sections from 119 patients with HPV- OSCC. Expression of ß2-microglobulin, MHC class I heavy chain, and large multifunctional peptidase 10 was quantified, and all data were correlated with patient outcome. We found that, consistent with previous reports, high numbers of CD8+ T cells at the invasive margin correlated significantly with prolonged overall survival (OS), while the number of FoxP3+ or PD-L1+ cells did not. Compiling the number of FoxP3+ or PD-L1+ cells within 30 µm of CD8+ T cells identified a significant association with a high number of suppressive elements close to CD8+ T cells and reduced OS. Integrating this information into a cumulative suppression index (CSI) increased correlation with OS. Incorporating tumor expression levels of APM components with CSI further improved prognostic power. This multiparametric immune profiling may be useful for stratifying patients with OSCC for clinical trials.

8.
Oncotarget ; 7(28): 44676-44685, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27172790

ABSTRACT

PURPOSE: To predict lymph node metastasis and prognosis in head and neck squamous cell carcinoma (HNSCC). RESULTS: The combination of membranous E-cadherin and membranous epidermal growth factor receptor (EGFR) quantified by QD technology with age, gender, and grade had greater predictive power than any of the single biomarkers or the two combined biomarkers quantified by conventional immunohistochemistry (IHC). The predictive power of this model was validated in another independent sample set; the predictive sensitivity of this model for LNM was 87.5%, with specificity up to 97.4%, and accuracy 92.9%. Furthermore, a higher membranous E-cadherin level was significantly correlated with better overall and disease-free survival (OS, DFS; P = 0.002, 0.033, respectively), while lower cytoplasmic vimentin and membranous EGFR levels were significantly correlated with better OS (P = 0.016 and 0.021, respectively). The combined biomarkers showed a stronger prognostic value for OS and DFS than any of the single biomarkers. METHODS: Multiplexed quantum dots (QDs) were used to simultaneously label E-cadherin, vimentin, and EGFR with ß-actin as an internal control. Primary tissue samples from 97 HNSCC patients, 49 with and 48 without LNM were included in the training set. Levels of membranous E-cadherin, cytoplasmic vimentin, and membranous EGFR were quantified by InForm software and correlated with clinical characteristics. CONCLUSIONS: Multiplexed subcellular QD quantification of EGFR and E-cadherin is a potential strategy for the prediction of LNM, DFS, and OS of HNSCC patients.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Quantum Dots , Aged , Cadherins/metabolism , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/therapy , ErbB Receptors/metabolism , Female , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/therapy , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Prognosis , Proportional Hazards Models , Sensitivity and Specificity , Vimentin/metabolism
9.
Oncoimmunology ; 4(10): e1022301, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26451293

ABSTRACT

Genomic profiling has identified several molecular oncodrivers in breast tumorigenesis. A thorough understanding of endogenous immune responses to these oncodrivers may provide insights into immune interventions for breast cancer (BC). We investigated systemic anti-HER2/neu CD4+ T-helper type-1 (Th1) responses in HER2-driven breast tumorigenesis. A highly significant stepwise Th1 response loss extending from healthy donors (HD), through HER2pos-DCIS, and ultimately to early stage HER2pos-invasive BC patients was detected by IFNγ ELISPOT. The anti-HER2 Th1 deficit was not attributable to host-level T-cell anergy, loss of immune competence, or increase in immunosuppressive phenotypes (Treg/MDSCs), but rather associated with a functional shift in IFNγ:IL-10-producing phenotypes. HER2high, but not HER2low, BC cells expressing IFNγ/TNF-α receptors were susceptible to Th1 cytokine-mediated apoptosis in vitro, which could be significantly rescued by neutralizing IFNγ and TNF-α, suggesting that abrogation of HER2-specific Th1 may reflect a mechanism of immune evasion in HER2-driven tumorigenesis. While largely unaffected by cytotoxic or HER2-targeted (trastuzumab) therapies, depressed Th1 responses in HER2pos-BC patients were significantly restored following HER2-pulsed dendritic cell (DC) vaccinations, suggesting that this Th1 defect is not "fixed" and can be corrected by immunologic interventions. Importantly, preserved anti-HER2 Th1 responses were associated with pathologic complete response to neoadjuvant trastuzumab/chemotherapy, while depressed responses were observed in patients incurring locoregional/systemic recurrence following trastuzumab/chemotherapy. Monitoring anti-HER2 Th1 reactivity following HER2-directed therapies may identify vulnerable subgroups at risk of clinicopathologic failure. In such patients, combinations of existing HER2-targeted therapies with strategies to boost anti-HER2 CD4+ Th1 immunity may decrease the risk of recurrence and thus warrant further investigation.

10.
J Immunother Cancer ; 3: 47, 2015.
Article in English | MEDLINE | ID: mdl-26500776

ABSTRACT

BACKGROUND: Adoptive T cell therapy (ACT) has shown great promise in melanoma, with over 50 % response rate in patients where autologous tumor-reactive tumor-infiltrating lymphocytes (TIL) can be cultured and expanded. A major limitation of ACT is the inability to generate or expand autologous tumor-reactive TIL in 25-45 % of patients tested. Methods that successfully identify tumors that are not suitable for TIL generation by standard methods would eliminate the costs of fruitless expansion and enable these patients to receive alternate therapy immediately. METHODS: Multispectral fluorescent immunohistochemistry with a panel including CD3, CD8, FoxP3, CD163, PD-L1 was used to analyze the tumor microenvironment in 17 patients with melanoma among our 36-patient cohort to predict successful TIL generation. Additionally, we compared tumor fragments and enzymatic digestion of tumor samples for efficiency in generating tumor-reactive TIL. RESULTS: Tumor-reactive TIL were generated from 21/36 (58 %) of melanomas and for 12/13 (92 %) tumors where both enzymatic and fragment methods were compared. TIL generation was successful in 10/13 enzymatic preparations and in 10/13 fragment cultures; combination of both methods resulted in successful generation of autologous tumor-reactive TIL in 12/13 patients. In 17 patients for whom tissue blocks were available, IHC analysis identified that while the presence of CD8(+) T cells alone was insufficient to predict successful TIL generation, the CD8(+) to FoxP3(+) ratio was predictive with a positive-predictive value (PPV) of 91 % and negative-predictive value (NPV) of 86 %. Incorporation of CD163+ macrophage numbers and CD8:PD-L1 ratio did not improve the PPV. However, the NPV could be improved to 100 % by including the ratio of CD8(+):PD-L1(+) expressing cells. CONCLUSION: This is the first study to apply 7-color multispectral immunohistochemistry to analyze the immune environment of tumors from patients with melanoma. Assessment of the data using unsupervised hierarchical clustering identified tumors from which we were unable to generate TIL. If substantiated, this immune profile could be applied to select patients for TIL generation. Additionally, this biomarker profile may also indicate a pre-existing immune response, and serve as a predictive biomarker of patients who will respond to checkpoint blockade. We postulate that expanding the spectrum of inhibitory cells and molecules assessed using this technique could guide combination immunotherapy treatments and improve response rates.

11.
Methods ; 70(1): 46-58, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25242720

ABSTRACT

Tissue sections offer the opportunity to understand a patient's condition, to make better prognostic evaluations and to select optimum treatments, as evidenced by the place pathology holds today in clinical practice. Yet, there is a wealth of information locked up in a tissue section that is only partially accessed, due mainly to the limitations of tools and methods. Often tissues are assessed primarily based on visual analysis of one or two proteins, or 2-3 DNA or RNA molecules. Even while analysis is still based on visual perception, image analysis is starting to address the variability of human perception. This is in contrast to measuring characteristics that are substantially out of reach of human perception, such as parameters revealed through co-expression, spatial relationships, heterogeneity, and low abundance molecules. What is not routinely accessed is the information revealed through simultaneous detection of multiple markers, the spatial relationships among cells and tissue in disease, and the heterogeneity now understood to be critical to developing effective therapeutic strategies. Our purpose here is to review and assess methods for multiplexed, quantitative, image analysis based approaches, using new multicolor immunohistochemistry methods, automated multispectral slide imaging, and advanced trainable pattern recognition software. A key aspect of our approach is presenting imagery in a workflow that engages the pathologist to utilize the strengths of human perception and judgment, while significantly expanding the range of metrics collectable from tissue sections and also provide a level of consistency and precision needed to support the complexities of personalized medicine.


Subject(s)
Immunohistochemistry/methods , Tyramine/chemistry , Animals , Automation , Biomarkers, Tumor , Breast Neoplasms/metabolism , DNA/chemistry , Female , Fluorescent Dyes/chemistry , Humans , Image Processing, Computer-Assisted/methods , Keratins/chemistry , Neoplasms/metabolism , Pattern Recognition, Automated , Perception , Precision Medicine , RNA/chemistry , Software
12.
Appl Immunohistochem Mol Morphol ; 22(5): 363-71, 2014.
Article in English | MEDLINE | ID: mdl-24162261

ABSTRACT

Detection of DNA mutations in tumor tissue can be a critical companion diagnostic test before prescription of a targeted therapy. Each method for detection of these mutations is associated with an analytic sensitivity that is a function of the percentage of tumor cells present in the specimen. Currently, tumor cell percentage is visually estimated resulting in an ordinal and highly variant result for a biologically continuous variable. We proposed that this aspect of DNA mutation testing could be standardized by developing a computer algorithm capable of accurately determining the percentage of malignant nuclei in an image of a hematoxylin and eosin-stained tissue. Using inForm software, we developed an algorithm, to calculate the percentage of malignant cells in histologic specimens of colon adenocarcinoma. A criterion standard was established by manually counting malignant and benign nuclei. Three pathologists also estimated the percentage of malignant nuclei in each image. Algorithm #9 had a median deviation from the criterion standard of 5.4% on the training set and 6.2% on the validation set. Compared with pathologist estimation, Algorithm #9 showed a similar ability to determine percentage of malignant nuclei. This method represents a potential future tool to assist in determining the percent of malignant nuclei present in a tissue section. Further validation of this algorithm or an improved algorithm may have value to more accurately assess percentage of malignant cells for companion diagnostic mutation testing.


Subject(s)
Adenocarcinoma/diagnosis , Cell Count/methods , Colonic Neoplasms/diagnosis , DNA/analysis , Mutation/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Algorithms , Automation, Laboratory , Carcinogenesis/genetics , Cell Count/standards , Cell Differentiation/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Diagnostic Errors/prevention & control , Humans , Observer Variation , Reference Standards , Software
13.
Cancer Cytopathol ; 121(3): 162-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22833451

ABSTRACT

BACKGROUND: Thyroid fine-needle aspiration (FNA) biopsy, the preoperative diagnostic standard of care for patients with thyroid nodules, has limitations. Spectral imaging captures visible light information that is beyond the capability of the human eye, potentially increasing the accuracy of FNA biopsy. In the current study, the authors demonstrated the feasibility of using spectral imaging in combination with automated spatial analysis based on trainable pattern recognition as an adjunct test for thyroid FNA classification by developing an algorithm that distinguishes between images of papillary thyroid carcinoma (PTC) and benign goiter (BG). METHODS: A multispectral camera was used to capture spectral images representing 100 cases of PTC and BG. Used in conjunction with commercial software, 10 cases were used as a training set to develop a "classifier," a classification algorithm that segments digitized multispectral images into regions of PTC, BG, and "nonfeature." This algorithm was used to generate a screening test and a diagnostic test that were validated on an independent set of images representing 30 cases of PTC and 30 cases of BG. RESULTS: The area under the receiver operating characteristic for the PTC/BG classifier was 0.90. The screening test had a sensitivity of 0.93 and a specificity of 0.73. The diagnostic test had a sensitivity of 0.70 and a specificity of 0.90. CONCLUSIONS: The authors developed image classification tests that distinguish between FNAs of PTC and BG, demonstrating the potential value of spatial spectral imaging as an adjunct test for the classification of thyroid FNA samples. The data support prospective testing to determine the value of the PTC/BG classifier in routine clinical use.


Subject(s)
Carcinoma, Papillary/classification , Goiter/classification , Image Interpretation, Computer-Assisted/instrumentation , Thyroid Neoplasms/classification , Thyroid Nodule/classification , Algorithms , Biopsy, Fine-Needle , Carcinoma, Papillary/pathology , Feasibility Studies , Goiter/pathology , Humans , Pilot Projects , ROC Curve , Thyroid Neoplasms/pathology , Thyroid Nodule/pathology , Validation Studies as Topic
14.
Endocrinology ; 153(4): 1673-83, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22355065

ABSTRACT

Islet amyloid is hypothesized to play a role in nonimmunologic transplanted islet graft loss. We performed a quantitative histologic analysis of liver biopsies from intrahepatic islet grafts transplanted in streptozotocin-induced diabetic cynomolgus macaques. Seven animals treated with antithymocyte globulin (ATG) and rapamycin or ATG and rituximab experienced islet graft rejection with lymphocytic infiltrates present on islet graft biopsies. Except for one case involving the oldest and largest donor where amyloid was present on initial biopsy 1 month after transplant, none of the six other cases with rejection contained amyloid, including one case biopsied serially to 25 months. In contrast, four out of six animals treated with ATG and rituximab and rapamycin had no evidence of rejection at the time of biopsy (two animals that discontinued rapamycin had mild periislet lymphocytes), and all four cases followed more than 4 months demonstrated amyloid deposition at subsequent time points. Amyloid severity increased with time after transplant (r = 0.68; P < 0.05) and with decreasing islet ß-cell area (r = -0.68; P < 0.05). In two islet recipients with no evidence of rejection and still normoglycemic and insulin independent at the first detection of amyloid, ß-cell secretory capacity declined over time coincident with increasing amyloid severity and decreasing ß-cell area, with both animals eventually becoming hyperglycemic and insulin dependent. Transplanted islet amyloid also developed in autologous islets placed sc. These results indicate that in cynomolgus macaques, transplanted islets may accumulate amyloid over time associated with subsequent decline in ß-cell mass and function and support the development of intrahepatic islet amyloid as a potential mechanism for nonimmunologic islet graft loss.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Disease Models, Animal , Islet Amyloid Polypeptide/metabolism , Islets of Langerhans Transplantation , Liver/metabolism , Macaca fascicularis/metabolism , Animals , Biopsy , Diabetes Mellitus, Experimental/chemically induced , Graft Rejection/etiology , Graft Rejection/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Liver/pathology , Pancreas/metabolism , Pancreas/pathology , Streptozocin/adverse effects , Time Factors
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.
Curr Protoc Mol Biol ; Chapter 14: Unit 14.19, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18972383

ABSTRACT

Combining images taken with light of specific wavelengths can dramatically enhance light-microscopic images. This technology is enabled by the availability of programmable filters that can be set to transmit light only of particular wavelengths. Spectral imaging technologies have become an important part of microscopy, and are particularly useful for analyzing samples that have been labeled with multiple (two or more) molecular markers. The most commonly used methodology for separating the markers from each other is linear unmixing, which results in a quantitative image of the location and amount of each marker present in the sample. The very complexity of these multilabel samples requires a high degree of sophistication in methods to visualize the results of unmixing. This article describes a wide range of useful visualization tools designed to better enable discrimination of different features in multilabeled tissue or cell samples. These commercially available tools can be attached to the standard laboratory light microscope to significantly enhance the power of light microscopy.


Subject(s)
Image Processing, Computer-Assisted/methods , Immunohistochemistry/methods , Laser Scanning Cytometry/methods , Microscopy, Fluorescence/methods , Animals , Fluorescent Dyes/analysis , Humans , Laser Scanning Cytometry/instrumentation , Microscopy, Fluorescence/instrumentation , Staining and Labeling/methods
17.
J Biomed Opt ; 10(4): 41207, 2005.
Article in English | MEDLINE | ID: mdl-16178631

ABSTRACT

The ability to image and quantitate fluorescently labeled markers in vivo has generally been limited by autofluorescence of the tissue. Skin, in particular, has a strong autofluorescence signal, particularly when excited in the blue or green wavelengths. Fluorescence labels with emission wavelengths in the near-infrared are more amenable to deep-tissue imaging, because both scattering and autofluorescence are reduced as wavelengths are increased, but even in these spectral regions, autofluorescence can still limit sensitivity. Multispectral imaging (MSI), however, can remove the signal degradation caused by autofluorescence while adding enhanced multiplexing capabilities. While the availability of spectral "libraries" makes multispectral analysis routine for well-characterized samples, new software tools have been developed that greatly simplify the application of MSI to novel specimens.


Subject(s)
Artifacts , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Microscopy, Fluorescence/methods , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Quantum Dots , Algorithms , Animals , Luminescent Proteins/metabolism , Male , Mice , Microscopy, Fluorescence/instrumentation , Phantoms, Imaging , Reproducibility of Results , Sensitivity and Specificity
18.
Biotechniques ; 39(6 Suppl): S33-7, 2005 Dec.
Article in English | MEDLINE | ID: mdl-20158502

ABSTRACT

Noninvasive in vivo imaging is a rapidly growing field with applications in basic biology, drug discovery and clinical medicine. Because of the high cost of magnetic resonance (MR)- and computed tomography (CT)-based systems, a great deal of effort has gone into developing optical imaging methods, which offer, in some modalities, the promise of high spatial resolution and the ability to detect multiple markers simultaneously However, the ability to image and quantitate fluorescently labeled tumors and other fluorescently labeled markers in vivo has generally been limited by the autofluorescence of the tissue, which reduces the sensitivity of detection and accuracy of quantitation of the labeled target. Multispectral imaging methodology, which spectrally characterizes and computationally eliminates autofluorescence, enhances signal-to-background dramatically, revealing otherwise invisible labeled targets. Signal-to-noise considerations can guide the choice of appropriate sensors for fluorescence-based imaging, which generally does not benefit from the use of highly cooled (and expensive) cameras. Effective use of spectral tools to remove autofluorescence signal requires accurate spectra of the individual components. Using manual and automated algorithms to generate these spectra, it is possible to detect as many as three fluorescent protein-labeled tumors and two separate autofluorescent signals in a single subject.


Subject(s)
Diagnostic Imaging/methods , Neoplasms/diagnosis , Spectrometry, Fluorescence/methods , Algorithms , Animals , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/analysis , Image Enhancement/methods , Mice , Mice, Nude , Proteins/analysis , Whole Body Imaging/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...