Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
J Immunother Cancer ; 8(2)2020 12.
Article En | MEDLINE | ID: mdl-33268350

Since the publication of the Society for Immunotherapy of Cancer's (SITC) original cancer immunotherapy biomarkers resource document, there have been remarkable breakthroughs in cancer immunotherapy, in particular the development and approval of immune checkpoint inhibitors, engineered cellular therapies, and tumor vaccines to unleash antitumor immune activity. The most notable feature of these breakthroughs is the achievement of durable clinical responses in some patients, enabling long-term survival. These durable responses have been noted in tumor types that were not previously considered immunotherapy-sensitive, suggesting that all patients with cancer may have the potential to benefit from immunotherapy. However, a persistent challenge in the field is the fact that only a minority of patients respond to immunotherapy, especially those therapies that rely on endogenous immune activation such as checkpoint inhibitors and vaccination due to the complex and heterogeneous immune escape mechanisms which can develop in each patient. Therefore, the development of robust biomarkers for each immunotherapy strategy, enabling rational patient selection and the design of precise combination therapies, is key for the continued success and improvement of immunotherapy. In this document, we summarize and update established biomarkers, guidelines, and regulatory considerations for clinical immune biomarker development, discuss well-known and novel technologies for biomarker discovery and validation, and provide tools and resources that can be used by the biomarker research community to facilitate the continued development of immuno-oncology and aid in the goal of durable responses in all patients.


Biomarkers, Tumor/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Humans
2.
J Cell Biochem ; 121(12): 4974-4990, 2020 Dec.
Article En | MEDLINE | ID: mdl-32692912

In preclinical cancer studies, three-dimensional (3D) cell spheroids and aggregates are preferred over monolayer cell cultures due to their architectural and functional similarity to solid tumors. We performed a proof-of-concept study to generate physiologically relevant and predictive preclinical models using non-small cell lung adenocarcinoma, and colon and colorectal adenocarcinoma cell line-derived 3D spheroids and aggregates. Distinct panels were designed to determine the expression profiles of frequently studied biomarkers of the two cancer subtypes. The lung adenocarcinoma panel included ALK, EGFR, TTF-1, and CK7 biomarkers, and the colon and colorectal adenocarcinoma panel included BRAF V600E, MSH2, MSH6, and CK20. Recent advances in immunofluorescence (IF) multiplexing and imaging technology enable simultaneous detection and quantification of multiple biomarkers on a single slide. In this study, we performed IF staining of multiple biomarkers per section on formalin-fixed paraffin-embedded 3D spheroids and aggregates. We optimized protocol parameters for automated IF and demonstrated staining concordance with automated chromogenic immunohistochemistry performed with validated protocols. Next, post-acquisition spectral unmixing of the captured fluorescent signals were utilized to delineate four differently stained biomarkers within a single multiplex IF image, followed by automated quantification of the expressed markers. This workflow has the potential to be adapted to preclinical high-throughput screening and drug efficacy studies utilizing 3D spheroids from cancer cell lines and patient-derived organoids. The process allows for cost, time, and resource savings through concurrent staining of several biomarkers on a single slide, the ability to study the interactions of multiple expressed proteins within a single region of interest, and enable quantitative assessment of biomarkers in cancer cells.

3.
Lab Invest ; 97(7): 873-885, 2017 07.
Article En | MEDLINE | ID: mdl-28504684

The ability to simultaneously visualize the presence, abundance, location and functional state of many targets in cells and tissues has been described as a true next-generation approach in immunohistochemistry (IHC). A typical requirement for multiplex IHC (mIHC) is the use of different animal species for each primary (1°Ab) and secondary (2°Ab) antibody pair. Although 1°Abs from different species have been used with differently labeled species-specific 2°Abs, quite often the appropriate combination of antibodies is not available. More recently, sequential detection of multiple antigens using 1°Abs from the same species used a microwaving treatment between successive antigen detection cycles to elute previously bound 1°Ab/2°Ab complex and therefore to prevent the cross-reactivity of anti-species 2°Abs used in subsequent detection cycles. We present here a fully automated 1°Ab/2°Ab complex heat deactivation (HD) method on Ventana's BenchMark ULTRA slide stainer. This method is applied to detection using fluorophore-conjugated tyramide deposited on the tissue and takes advantage of the strong covalent bonding of the detection substrate to the tissue, preventing its elution in the HD process. The HD process was characterized for (1) effectiveness in preventing Ab cross-reactivity, (2) impact on the epitopes and (3) impact on the fluorophores. An automated 5-plex fluorescent IHC assay was further developed using the HD method and rabbit 1°Abs for CD3, CD8, CD20, CD68 and FoxP3 immune biomarkers in human tissue specimens. The fluorophores were carefully chosen and the narrow-band filters were designed to allow visualization of the staining under fluorescent microscope with minimal bleed through. The automated 5-plex fluorescent IHC assay achieved staining results comparable to the respective single-plex chromogenic IHC assays. This technology enables automated mIHC using unmodified 1°Abs from same species and the corresponding anti-species 2°Ab on a clinically established automated platform to ensure staining quality, reliability and reproducibility.


Amides/chemistry , Antibodies/chemistry , Fluorescent Dyes/chemistry , Image Processing, Computer-Assisted/methods , Immunohistochemistry/methods , Amides/metabolism , Antibodies/metabolism , Breast/chemistry , Female , Fluorescent Dyes/metabolism , Humans , Neoplasms/chemistry , Palatine Tonsil/chemistry , Reproducibility of Results
4.
Radiology ; 280(3): 826-36, 2016 09.
Article En | MEDLINE | ID: mdl-27332865

Purpose To quantitatively determine the limit of detection of marrow stromal cells (MSC) after cardiac cell therapy (CCT) in swine by using clinical positron emission tomography (PET) reporter gene imaging and magnetic resonance (MR) imaging with cell prelabeling. Materials and Methods Animal studies were approved by the institutional administrative panel on laboratory animal care. Seven swine received 23 intracardiac cell injections that contained control MSC and cell mixtures of MSC expressing a multimodality triple fusion (TF) reporter gene (MSC-TF) and bearing superparamagnetic iron oxide nanoparticles (NP) (MSC-TF-NP) or NP alone. Clinical MR imaging and PET reporter gene molecular imaging were performed after intravenous injection of the radiotracer fluorine 18-radiolabeled 9-[4-fluoro-3-(hydroxyl methyl) butyl] guanine ((18)F-FHBG). Linear regression analysis of both MR imaging and PET data and nonlinear regression analysis of PET data were performed, accounting for multiple injections per animal. Results MR imaging showed a positive correlation between MSC-TF-NP cell number and dephasing (dark) signal (R(2) = 0.72, P = .0001) and a lower detection limit of at least approximately 1.5 × 10(7) cells. PET reporter gene imaging demonstrated a significant positive correlation between MSC-TF and target-to-background ratio with the linear model (R(2) = 0.88, P = .0001, root mean square error = 0.523) and the nonlinear model (R(2) = 0.99, P = .0001, root mean square error = 0.273) and a lower detection limit of 2.5 × 10(8) cells. Conclusion The authors quantitatively determined the limit of detection of MSC after CCT in swine by using clinical PET reporter gene imaging and clinical MR imaging with cell prelabeling. (©) RSNA, 2016 Online supplemental material is available for this article.


Genes, Reporter , Heart/diagnostic imaging , Mesenchymal Stem Cell Transplantation , Molecular Imaging/methods , Multimodal Imaging/methods , Animals , Fluorine Radioisotopes , Guanine/analogs & derivatives , Magnetic Resonance Imaging , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Swine
5.
Radiology ; 280(3): 815-25, 2016 09.
Article En | MEDLINE | ID: mdl-27308957

Purpose To use multimodality reporter-gene imaging to assess the serial survival of marrow stromal cells (MSC) after therapy for myocardial infarction (MI) and to determine if the requisite preclinical imaging end point was met prior to a follow-up large-animal MSC imaging study. Materials and Methods Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care. Mice (n = 19) that had experienced MI were injected with bone marrow-derived MSC that expressed a multimodality triple fusion (TF) reporter gene. The TF reporter gene (fluc2-egfp-sr39ttk) consisted of a human promoter, ubiquitin, driving firefly luciferase 2 (fluc2), enhanced green fluorescent protein (egfp), and the sr39tk positron emission tomography reporter gene. Serial bioluminescence imaging of MSC-TF and ex vivo luciferase assays were performed. Correlations were analyzed with the Pearson product-moment correlation, and serial imaging results were analyzed with a mixed-effects regression model. Results Analysis of the MSC-TF after cardiac cell therapy showed significantly lower signal on days 8 and 14 than on day 2 (P = .011 and P = .001, respectively). MSC-TF with MI demonstrated significantly higher signal than MSC-TF without MI at days 4, 8, and 14 (P = .016). Ex vivo luciferase activity assay confirmed the presence of MSC-TF on days 8 and 14 after MI. Conclusion Multimodality reporter-gene imaging was successfully used to assess serial MSC survival after therapy for MI, and it was determined that the requisite preclinical imaging end point, 14 days of MSC survival, was met prior to a follow-up large-animal MSC study. (©) RSNA, 2016 Online supplemental material is available for this article.


Genes, Reporter , Mesenchymal Stem Cell Transplantation/methods , Molecular Imaging , Multimodal Imaging , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/therapy , Animals , Female , Luciferases, Firefly/metabolism , Luminescent Measurements , Mice , Mice, Nude , Positron-Emission Tomography , Transfection
6.
PLoS One ; 10(11): e0142665, 2015.
Article En | MEDLINE | ID: mdl-26569397

OBJECTIVE: Ultra-small superparamagnetic iron oxide nanoparticles (USPIO) are promising contrast agents for magnetic resonance imaging (MRI). USPIO mediated proton relaxation rate enhancement is strongly dependent on compartmentalization of the agent and can vary depending on their intracellular or extracellular location in the tumor microenvironment. We compared the T1- and T2-enhancement pattern of intracellular and extracellular USPIO in mouse models of cancer and pilot data from patients. A better understanding of these MR signal effects will enable non-invasive characterizations of the composition of the tumor microenvironment. MATERIALS AND METHODS: Six 4T1 and six MMTV-PyMT mammary tumors were grown in mice and imaged with ferumoxytol-enhanced MRI. R1 relaxation rates were calculated for different tumor types and different tumor areas and compared with histology. The transendothelial leakage rate of ferumoxytol was obtained by our measured relaxivity of ferumoxytol and compared between different tumor types, using a t-test. Additionally, 3 patients with malignant sarcomas were imaged with ferumoxytol-enhanced MRI. T1- and T2-enhancement patterns were compared with histopathology in a descriptive manner as a proof of concept for clinical translation of our observations. RESULTS: 4T1 tumors showed central areas of high signal on T1 and low signal on T2 weighted MR images, which corresponded to extracellular nanoparticles in a necrotic core on histopathology. MMTV-PyMT tumors showed little change on T1 but decreased signal on T2 weighted images, which correlated to compartmentalized nanoparticles in tumor associated macrophages. Only 4T1 tumors demonstrated significantly increased R1 relaxation rates of the tumor core compared to the tumor periphery (p<0.001). Transendothelial USPIO leakage was significantly higher for 4T1 tumors (3.4±0.9x10-3 mL/min/100cm3) compared to MMTV-PyMT tumors (1.0±0.9x10-3 mL/min/100 cm3). Likewise, ferumoxytol imaging in patients showed similar findings with high T1 signal in areas of tumor necrosis and low signal in areas of intracellularly compartmentalized iron. CONCLUSION: Differential T1- and T2-enhancement patterns of USPIO in tumors enable conclusions about their intracellular and extracellular location. This information can be used to characterize the composition of the tumor microenvironment.


Bone Neoplasms/pathology , Ferric Compounds/chemistry , Ferrosoferric Oxide/chemistry , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Metal Nanoparticles/chemistry , Neoplasms/pathology , Osteosarcoma/pathology , Sarcoma, Ewing/pathology , Animals , Contrast Media/chemistry , Diagnostic Imaging , Disease Models, Animal , Female , Humans , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Mice, Transgenic , Necrosis , Neoplasm Transplantation , Pilot Projects
7.
Mol Imaging Biol ; 16(6): 865-76, 2014 Dec.
Article En | MEDLINE | ID: mdl-24845530

PURPOSE: Human pluripotency gene networks (PGNs), controlled in part by Oct4, are central to understanding pluripotent stem cells, but current fluorescent reporter genes (RGs) preclude noninvasive assessment of Oct4 dynamics in living subjects. PROCEDURES: To assess Oc4 activity noninvasively, we engineered a mouse embryonic stem cell line which encoded both a pOct4-hrluc (humanized renilla luciferase) reporter and a pUbi-hfluc2-gfp (humanized firefly luciferase 2 fused to green fluorescent protein) reporter. RESULTS: In cell culture, pOct4-hRLUC activity demonstrated a peak at 48 h (day 2) and significant downregulation by 72 h (day 3) (p=0.0001). Studies in living subjects demonstrated significant downregulation in pOct4-hRLUC activity between 12 and 144 h (p = 0.001) and between 12 and 168 h (p = 0.0003). pOct4-hRLUC signal dynamics after implantation was complex, characterized by transient upregulation after initial downregulation in all experiments (n = 10, p = 0.01). As expected, cell culture differentiation of the engineered mouse embryonic stem cell line demonstrated activation of mesendodermal, mesodermal, endodermal, and ectodermal master regulators of differentiation, indicating potency to form all three germ layers. CONCLUSIONS: We conclude that the Oct4-hrluc RG system enables noninvasive Oct4 imaging in cell culture and in living subjects.


Embryonic Stem Cells/physiology , Molecular Imaging/methods , Octamer Transcription Factor-3/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Embryonic Stem Cells/cytology , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Microscopy, Fluorescence , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells , Rats
8.
Contrast Media Mol Imaging ; 8(3): 281-8, 2013.
Article En | MEDLINE | ID: mdl-23606432

Tumor-associated macrophages (TAM) maintain a chronic inflammation in cancers, which is associated with tumor aggressiveness and poor prognosis. The purpose of this study was to: (1) evaluate the pharmacokinetics and tolerability of the novel ultrasmall superparamagnetic iron oxide nanoparticle (USPIO) compound GEH121333; (2) assess whether GEH121333 can serve as a MR imaging biomarker for TAM; and (3) compare tumor MR enhancement profiles between GEH121333 and ferumoxytol. Blood half-lives of GEH121333 and ferumoxytol were measured by relaxometry (n = 4 each). Tolerance was assessed in healthy rats injected with high dose GEH121333, vehicle or saline (n = 4 each). Animals were monitored for 7 days regarding body weight, complete blood counts and serum chemistry, followed by histological evaluation of visceral organs. MR imaging was performed on mice harboring MMTV-PyMT-derived breast adenocarcinomas using a 7 T scanner before and up to 72 h post-injection (p.i.) of GEH121333 (n = 10) or ferumoxytol (n = 9). Tumor R1, R2* relaxation rates were compared between different experimental groups and time points, using a linear mixed effects model with a random effect for each animal. MR data were correlated with histopathology. GEH121333 showed a longer circulation half-life than ferumoxytol. Intravenous GEH121333 did not produce significant adverse effects in rats. All tumors demonstrated significant enhancement on T1, T2 and T2*-weighted images at 1, 24, 48 and 72 h p.i. GEH121333 generated stronger tumor T2* enhancement than ferumoxytol. Histological analysis verified intracellular compartmentalization of GEH121333 by TAM at 24, 48 and 72 h p.i. MR imaging with GEH121333 nanoparticles represents a novel biomarker for TAM assessment. This new USPIO MR contrast agent provides a longer blood half-life and better TAM enhancement compared with the iron supplement ferumoxytol.


Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Contrast Media/chemical synthesis , Dextrans/pharmacokinetics , Macrophages/immunology , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Animals , Cell Line, Tumor , Contrast Media/pharmacokinetics , Dextrans/chemical synthesis , Macrophages/pathology , Metabolic Clearance Rate , Mice , Organ Specificity , Rats , Rats, Inbred Lew , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
9.
Curr Pharm Biotechnol ; 12(4): 497-507, 2011 Apr.
Article En | MEDLINE | ID: mdl-21342105

Gene-directed enzyme prodrug therapy (GDEPT) is one of the promising alternatives to conventional chemotherapy. Suicide gene therapy based anticancer strategy involves selective introduction of a foreign gene into tumor cells to produce a foreign enzyme that can activate an inert prodrug to its cytotoxic form and cause tumor cell death. In this review, we present three most promising suicide gene/prodrug combinations (1) herpes simplex virus thymidine kinase (HSV-TK) with ganciclovir (GCV), (2) cytosine deaminase (CD) from bacteria or yeast with 5-fluorocytodine (5-FC) and (3) bacterial nitroreductase (NTR) with 5-(azaridin-1-yl)-2,4-dinitrobenzamide (CB1954) and discuss how molecular imaging may improve therapy strategies. Current advances in noninvasive imaging technologies can measure vector dose, tumor selectivity, transgene expression and biodistribution of therapeutic gene with the aid of reporter genes and imageable probes from live animal. In this review we will discuss various imaging modalities - Optical, Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT), and highlight some of the approaches that can advance prodrug cancer therapy from bench to clinic.


Antineoplastic Agents/therapeutic use , Cytosine Deaminase/genetics , Genetic Therapy/methods , Nitroreductases/genetics , Prodrugs/therapeutic use , Thymidine Kinase/genetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Bystander Effect , Cell Death , Gene Transfer Techniques , Genes, Reporter , Genes, Transgenic, Suicide , Humans , Molecular Imaging/methods , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology , Neoplasms/therapy , Prodrugs/administration & dosage , Prodrugs/chemistry
10.
J Biomed Opt ; 15(2): 020509, 2010.
Article En | MEDLINE | ID: mdl-20459220

Fluorescence image-guided surgery (FIGS) is an emerging technique in oncology, neurology, and cardiology. To adapt intraoperative imaging for various surgical applications, increasingly flexible and compact FIGS instruments are necessary. We present a compact, portable FIGS system and demonstrate its use in cardiovascular mapping in a preclinical model of myocardial ischemia. Our system uses fiber optic delivery of laser diode excitation, custom optics with high collection efficiency, and compact consumer-grade cameras as a low-cost and compact alternative to open surgical FIGS systems. Dramatic size and weight reduction increases flexibility and access, and allows for handheld use or unobtrusive positioning over the surgical field.


Fiber Optic Technology/instrumentation , Image Enhancement/instrumentation , Microscopy, Fluorescence/instrumentation , Myocardial Ischemia/pathology , Myocardial Ischemia/surgery , Surgery, Computer-Assisted/instrumentation , Animals , Computer-Aided Design , Equipment Design , Equipment Failure Analysis , Male , Miniaturization , Rats , Rats, Wistar , Reproducibility of Results , Sensitivity and Specificity
11.
J Biomed Opt ; 14(6): 060504, 2009.
Article En | MEDLINE | ID: mdl-20059235

We describe a wide-field preclinical imaging system optimized for time-gated detection of quantum dot fluorescence emission. As compared to continuous wave measurements, image contrast was substantially improved by suppression of short-lifetime background autofluorescence. Real-time (8 frames/s) biological imaging of subcutaneous quantum dot injections is demonstrated simultaneously in multiple living mice.


Fluorescent Dyes/chemistry , Quantum Dots , Whole Body Imaging/methods , Animals , Fluorescence , Fluorescent Dyes/administration & dosage , Mice , Mice, Nude , Whole Body Imaging/instrumentation
12.
Eur J Nucl Med Mol Imaging ; 35(12): 2275-85, 2008 Dec.
Article En | MEDLINE | ID: mdl-18661130

PURPOSE: Firefly luciferase catalyzes the oxidative decarboxylation of D: -luciferin to oxyluciferin in the presence of cofactors, producing bioluminescence. This reaction is used in optical bioluminescence-based molecular imaging approaches to detect the expression of the firefly luciferase reporter gene. Biokinetics and distribution of the substrate most likely have a significant impact on levels of light signal and therefore need to be investigated. METHODS: Benzene ring (14)C(U)-labeled D-luciferin was utilized. Cell uptake and efflux assays, murine biodistribution, autoradiography and CCD-camera based optical bioluminescence imaging were carried out to examine the in vitro and in vivo characteristics of the tracer in cell culture and in living mice respectively. RESULTS: Radiolabeled and unlabeled D-luciferin revealed comparable levels of light emission when incubated with equivalent amounts of the firefly luciferase enzyme. Cell uptake assays in pCMV-luciferase-transfected cells showed slow trapping of the tracer and relatively low uptake values (up to 22.9-fold higher in firefly luciferase gene-transfected vs. nontransfected cells, p = 0.0002). Biodistribution studies in living mice after tail-vein injection of (14)C-D-luciferin demonstrated inhomogeneous tracer distribution with early predominant high radioactivity levels in kidneys (10.6% injected dose [ID]/g) and liver (11.9% ID/g), followed at later time points by the bladder (up to 81.3% ID/g) and small intestine (6.5% ID/g), reflecting the elimination routes of the tracer. Kinetics and uptake levels profoundly differed when using alternate injection routes (intravenous versus intraperitoneal). No clear trapping of (14)C-D-luciferin in firefly luciferase-expressing tissues could be observed in vivo. CONCLUSIONS: The data obtained with (14)C-D-luciferin provide insights into the dynamics of D: -luciferin cell uptake, intracellular accumulation, and efflux. Results of the biodistribution and autoradiographic studies should be useful for optimizing and adapting optical imaging protocols to specific experimental settings when utilizing the firefly luciferase and D: -luciferin system.


Benzothiazoles/metabolism , Benzothiazoles/pharmacokinetics , Genes, Reporter , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Luminescent Measurements , Adenoviridae/genetics , Animals , Autoradiography , Benzothiazoles/administration & dosage , Benzothiazoles/chemistry , Biocatalysis , Biological Transport/drug effects , Buffers , Carbon Radioisotopes/chemistry , Cattle , Cell Line , Cold Temperature , Culture Media , Gene Expression , Humans , Injections , Intracellular Space/metabolism , Kidney/metabolism , Kinetics , Tissue Distribution
13.
Lab Anim (NY) ; 36(8): 40-3, 2007 Sep.
Article En | MEDLINE | ID: mdl-17721532

As small-animal fluorescence imaging becomes increasingly accessible to a broad spectrum of users, many lab animal researchers are just beginning to be exposed to its challenges. One setback to fluorescence imaging is background autofluorescence generated in animal tissue and in ingested food. The authors bring this issue into focus, and show how autofluorescence can be reduced in nude mice through selection of appropriate excitation wavelength and mouse diet.


Artifacts , Fluorescence , Laboratory Animal Science/methods , Microscopy, Fluorescence, Multiphoton/methods , Spectrometry, Fluorescence/methods , Animal Feed , Animals , Diet/standards , Female , Food, Formulated , Lasers , Mice , Mice, Nude
...