Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Publication year range
1.
Cell ; 183(3): 818-834.e13, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33038342

ABSTRACT

Many approaches to identify therapeutically relevant neoantigens couple tumor sequencing with bioinformatic algorithms and inferred rules of tumor epitope immunogenicity. However, there are no reference data to compare these approaches, and the parameters governing tumor epitope immunogenicity remain unclear. Here, we assembled a global consortium wherein each participant predicted immunogenic epitopes from shared tumor sequencing data. 608 epitopes were subsequently assessed for T cell binding in patient-matched samples. By integrating peptide features associated with presentation and recognition, we developed a model of tumor epitope immunogenicity that filtered out 98% of non-immunogenic peptides with a precision above 0.70. Pipelines prioritizing model features had superior performance, and pipeline alterations leveraging them improved prediction performance. These findings were validated in an independent cohort of 310 epitopes prioritized from tumor sequencing data and assessed for T cell binding. This data resource enables identification of parameters underlying effective anti-tumor immunity and is available to the research community.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes/immunology , Neoplasms/immunology , Alleles , Antigen Presentation/immunology , Cohort Studies , Humans , Peptides/immunology , Programmed Cell Death 1 Receptor , Reproducibility of Results
2.
Nature ; 615(7953): 687-696, 2023 03.
Article in English | MEDLINE | ID: mdl-36356599

ABSTRACT

T cell receptors (TCRs) enable T cells to specifically recognize mutations in cancer cells1-3. Here we developed a clinical-grade approach based on CRISPR-Cas9 non-viral precision genome-editing to simultaneously knockout the two endogenous TCR genes TRAC (which encodes TCRα) and TRBC (which encodes TCRß). We also inserted into the TRAC locus two chains of a neoantigen-specific TCR (neoTCR) isolated from circulating T cells of patients. The neoTCRs were isolated using a personalized library of soluble predicted neoantigen-HLA capture reagents. Sixteen patients with different refractory solid cancers received up to three distinct neoTCR transgenic cell products. Each product expressed a patient-specific neoTCR and was administered in a cell-dose-escalation, first-in-human phase I clinical trial ( NCT03970382 ). One patient had grade 1 cytokine release syndrome and one patient had grade 3 encephalitis. All participants had the expected side effects from the lymphodepleting chemotherapy. Five patients had stable disease and the other eleven had disease progression as the best response on the therapy. neoTCR transgenic T cells were detected in tumour biopsy samples after infusion at frequencies higher than the native TCRs before infusion. This study demonstrates the feasibility of isolating and cloning multiple TCRs that recognize mutational neoantigens. Moreover, simultaneous knockout of the endogenous TCR and knock-in of neoTCRs using single-step, non-viral precision genome-editing are achieved. The manufacture of neoTCR engineered T cells at clinical grade, the safety of infusing up to three gene-edited neoTCR T cell products and the ability of the transgenic T cells to traffic to the tumours of patients are also demonstrated.


Subject(s)
Cell- and Tissue-Based Therapy , Gene Editing , Neoplasms , Precision Medicine , Receptors, Antigen, T-Cell , T-Lymphocytes , Transgenes , Humans , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biopsy , Cell- and Tissue-Based Therapy/adverse effects , Cell- and Tissue-Based Therapy/methods , Cytokine Release Syndrome/complications , Disease Progression , Encephalitis/complications , Gene Knock-In Techniques , Gene Knockout Techniques , Genes, T-Cell Receptor alpha , Genes, T-Cell Receptor beta , Mutation , Neoplasms/complications , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Patient Safety , Precision Medicine/adverse effects , Precision Medicine/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transgenes/genetics , HLA Antigens/immunology , CRISPR-Cas Systems
3.
Proc Natl Acad Sci U S A ; 116(44): 22014-22019, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611413

ABSTRACT

T cells can be controllably stimulated through antigen-specific or nonspecific protocols. Accompanying functional hallmarks of T cell activation can include cytoskeletal reorganization, cell size increase, and cytokine secretion. Photon-induced near-field electron microscopy (PINEM) is used to image and quantify evanescent electric fields at the surface of T cells as a function of various stimulation conditions. While PINEM signal strength scales with multiple of the biophysical changes associated with T cell functional activation, it mostly strongly correlates with antigen-engagement of the T cell receptors, even under conditions that do not lead to functional T cell activation. PINEM image analysis suggests that a stimulation-induced reorganization of T cell surface structure, especially over length scales of a few hundred nanometers, is the dominant contributor to these PINEM signal changes. These experiments reveal that PINEM can provide a sensitive label-free probe of nanoscale cellular surface structures.


Subject(s)
Lymphocyte Activation , T-Lymphocytes/ultrastructure , Humans , Jurkat Cells , Microscopy, Electron/methods , Surface Properties
4.
PLoS Comput Biol ; 15(6): e1007034, 2019 06.
Article in English | MEDLINE | ID: mdl-31166947

ABSTRACT

Phenotypic plasticity is associated with non-genetic drug tolerance in several cancers. Such plasticity can arise from chromatin remodeling, transcriptomic reprogramming, and/or protein signaling rewiring, and is characterized as a cell state transition in response to molecular or physical perturbations. This, in turn, can confound interpretations of drug responses and resistance development. Using BRAF-mutant melanoma cell lines as the prototype, we report on a joint theoretical and experimental investigation of the cell-state transition dynamics associated with BRAF inhibitor drug tolerance. Thermodynamically motivated surprisal analysis of transcriptome data was used to treat the cell population as an entropy maximizing system under the influence of time-dependent constraints. This permits the extraction of an epigenetic potential landscape for drug-induced phenotypic evolution. Single-cell flow cytometry data of the same system were modeled with a modified Fokker-Planck-type kinetic model. The two approaches yield a consistent picture that accounts for the phenotypic heterogeneity observed over the course of drug tolerance development. The results reveal that, in certain plastic cancers, the population heterogeneity and evolution of cell phenotypes may be understood by accounting for the competing interactions of the epigenetic potential landscape and state-dependent cell proliferation. Accounting for such competition permits accurate, experimentally verifiable predictions that can potentially guide the design of effective treatment strategies.


Subject(s)
Drug Resistance, Neoplasm , Evolution, Molecular , Melanoma , Phenotype , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Melanoma/genetics , Melanoma/physiopathology , Models, Biological , Transcriptome/drug effects , Transcriptome/genetics
5.
Annu Rev Biomed Eng ; 17: 91-112, 2015.
Article in English | MEDLINE | ID: mdl-26643019

ABSTRACT

Digital microfluidics (DMF) is a droplet-based liquid-handling technology that has recently become popular for cell culture and analysis. In DMF, picoliter- to microliter-sized droplets are manipulated on a planar surface using electric fields, thus enabling software-reconfigurable operations on individual droplets, such as move, merge, split, and dispense from reservoirs. Using this technique, multistep cell-based processes can be carried out using simple and compact instrumentation, making DMF an attractive platform for eventual integration into routine biology workflows. In this review, we summarize the state-of-the-art in DMF cell culture, and describe design considerations, types of DMF cell culture, and cell-based applications of DMF.


Subject(s)
Cell Culture Techniques/methods , Microfluidic Analytical Techniques/methods , Adsorption , Cell Culture Techniques/instrumentation , Cell Separation , Drug Evaluation, Preclinical , Electrowetting , Humans , Lab-On-A-Chip Devices , Materials Testing , Poloxamer , Proteins/chemistry , Surface-Active Agents
6.
Anal Chem ; 87(7): 3902-10, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25759941

ABSTRACT

There is great interest in the development of integrated tools allowing for miniaturized sample processing, including solid phase extraction (SPE). We introduce a new format for microfluidic SPE relying on C18-functionalized magnetic beads that can be manipulated in droplets in a digital microfluidic platform. This format provides the opportunity to tune the amount (and potentially the type) of stationary phase on-the-fly, and allows the removal of beads after the extraction (to enable other operations in same device-space), maintaining device reconfigurability. Using the new method, we employed a design of experiments (DOE) operation to enable automated on-chip optimization of elution solvent composition for reversed phase SPE of a model system. Further, conditions were selected to enable on-chip fractionation of multiple analytes. Finally, the method was demonstrated to be useful for online cleanup of extracts from dried blood spot (DBS) samples. We anticipate this combination of features will prove useful for separating a wide range of analytes, from small molecules to peptides, from complex matrices.


Subject(s)
Aminoquinolines/analysis , Angiotensin I/analysis , Dried Blood Spot Testing , Magnetite Nanoparticles/chemistry , Microfluidic Analytical Techniques , Solvents/chemistry , Equipment Design , Humans , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Solid Phase Extraction
7.
Clin Chem ; 61(2): 420-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25512641

ABSTRACT

BACKGROUND: Whereas disease surveillance for infectious diseases such as rubella is important, it is critical to identify pregnant women at risk of passing rubella to their offspring, which can be fatal and can result in congenital rubella syndrome (CRS). The traditional centralized model for diagnosing rubella is cost-prohibitive in resource-limited settings, representing a major obstacle to the prevention of CRS. As a step toward decentralized diagnostic systems, we developed a proof-of-concept digital microfluidic (DMF) diagnostic platform that possesses the flexibility and performance of automated immunoassay platforms used in central facilities, but with a form factor the size of a shoebox. METHODS: DMF immunoassays were developed with integrated sample preparation for the detection of rubella virus (RV) IgG and IgM. The performance (sensitivity and specificity) of the assays was evaluated with serum and plasma samples from a commercial antirubella mixed-titer performance panel. RESULTS: The new platform performed the essential processing steps, including sample aliquoting for 4 parallel assays, sample dilution, and IgG blocking. Testing of performance panel samples yielded diagnostic sensitivity and specificity of 100% and 100% for both RV IgG and RV IgM. With 1.8 µL sample per assay, 4 parallel assays were performed in approximately 30 min with <10% mean CV. CONCLUSIONS: This proof of concept establishes DMF-powered immunoassays as being potentially useful for the diagnosis of infectious disease.


Subject(s)
Antibodies, Viral/blood , Immunoassay/instrumentation , Immunoglobulin G/blood , Immunoglobulin M/blood , Microfluidic Analytical Techniques/instrumentation , Rubella virus/immunology , Rubella/diagnosis , Antibodies, Viral/immunology , Equipment Design , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Pregnancy , Rubella/blood , Rubella/immunology , Rubella virus/isolation & purification , Sensitivity and Specificity
8.
Anal Chem ; 86(16): 8466-72, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25058398

ABSTRACT

Many important biomarkers for disease diagnosis are present at low concentrations in human serum. These biomarkers are masked in proteomic analysis by highly abundant proteins such as human serum albumin (HSA) and immunoglobulins (IgGs) which account for up to 80% of the total protein content of serum. Traditional depletion methods using macro-scale LC-columns for highly abundant proteins involve slow separations which impart considerable dilution to the samples. Furthermore, most techniques lack the ability to process multiple samples simultaneously. We present a method of protein depletion using superparamagnetic beads coated in anti-HSA, Protein A, and Protein G, manipulated by digital microfluidics (DMF). The depletion process was capable of up to 95% protein depletion efficiency for IgG and HSA in 10 min for four samples simultaneously, which resulted in an approximately 4-fold increase in signal-to-noise ratio in MALDI-MS analysis for a low abundance protein, hemopexin. This rapid and automated method has the potential to greatly improve the process of biomarker identification.


Subject(s)
Antibodies, Immobilized/chemistry , Blood Proteins/isolation & purification , Immunomagnetic Separation/instrumentation , Microfluidic Analytical Techniques/instrumentation , Equipment Design , Humans , Magnets/chemistry , Proteomics/instrumentation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/instrumentation
9.
Anal Chem ; 85(20): 9638-46, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23978190

ABSTRACT

We introduce an automated digital microfluidic (DMF) platform capable of performing immunoassays from sample to analysis with minimal manual intervention. This platform features (a) a 90 Pogo pin interface for digital microfluidic control, (b) an integrated (and motorized) photomultiplier tube for chemiluminescent detection, and (c) a magnetic lens assembly which focuses magnetic fields into a narrow region on the surface of the DMF device, facilitating up to eight simultaneous digital microfluidic magnetic separations. The new platform was used to implement a three-level full factorial design of experiments (DOE) optimization for thyroid-stimulating hormone immunoassays, varying (1) the analyte concentration, (2) the sample incubation time, and (3) the sample volume, resulting in an optimized protocol that reduced the detection limit and sample incubation time by up to 5-fold and 2-fold, respectively, relative to those from previous work. To our knowledge, this is the first report of a DOE optimization for immunoassays in a microfluidic system of any format. We propose that this new platform paves the way for a benchtop tool that is useful for implementing immunoassays in near-patient settings, including community hospitals, physicians' offices, and small clinical laboratories.

10.
Cell Rep ; 42(10): 113212, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37792533

ABSTRACT

Local immune activation at mucosal surfaces, mediated by mucosal lymphoid tissues, is vital for effective immune responses against pathogens. While pathogens like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can spread to multiple organs, patients with coronavirus disease 2019 (COVID-19) primarily experience inflammation and damage in their lungs. To investigate this apparent organ-specific immune response, we develop an analytical framework that recognizes the significance of mucosal lymphoid tissues. This framework combines histology, immunofluorescence, spatial transcript profiling, and mathematical modeling to identify cellular and gene expression differences between the lymphoid tissues of the lung and the gut and predict the determinants of those differences. Our findings indicate that mucosal lymphoid tissues are pivotal in organ-specific immune response to SARS-CoV-2, mediating local inflammation and tissue damage and contributing to immune dysfunction. The framework developed here has potential utility in the study of long COVID and may streamline biomarker discovery and treatment design for diseases with differential pathologies at the organ level.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Post-Acute COVID-19 Syndrome , Inflammation , Immunity
11.
Commun Biol ; 6(1): 528, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37193826

ABSTRACT

The discovery and characterization of antigen-specific CD8+ T cell clonotypes typically involves the labor-intensive synthesis and construction of peptide-MHC tetramers. We adapt single-chain trimer (SCT) technologies into a high throughput platform for pMHC library generation, showing that hundreds can be rapidly prepared across multiple Class I HLA alleles. We use this platform to explore the impact of peptide and SCT template mutations on protein expression yield, thermal stability, and functionality. SCT libraries were an efficient tool for identifying T cells recognizing commonly reported viral epitopes. We then construct SCT libraries to capture SARS-CoV-2 specific CD8+ T cells from COVID-19 participants and healthy donors. The immunogenicity of these epitopes is validated by functional assays of T cells with cloned TCRs captured using SCT libraries. These technologies should enable the rapid analyses of peptide-based T cell responses across several contexts, including autoimmunity, cancer, or infectious disease.


Subject(s)
CD8-Positive T-Lymphocytes , COVID-19 , Humans , SARS-CoV-2/genetics , Antigens , Epitopes , Peptides/genetics
12.
Anal Chem ; 84(20): 8805-12, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23013543

ABSTRACT

We introduce a new format for particle-based immunoassays relying on digital microfluidics (DMF) and magnetic forces to separate and resuspend antibody-coated paramagnetic particles. In DMF, fluids are electrostatically controlled as discrete droplets (picoliters to microliters) on an array of insulated electrodes. By applying appropriate sequences of potentials to these electrodes, multiple droplets can be manipulated simultaneously and various droplet operations can be achieved using the same device design. This flexibility makes DMF well-suited for applications that require complex, multistep protocols such as immunoassays. Here, we report the first particle-based immunoassay on DMF without the aid of oil carrier fluid to enable droplet movement (i.e., droplets are surrounded by air instead of oil). This new format allowed the realization of a novel on-chip particle separation and resuspension method capable of removing greater than 90% of unbound reagents in one step. Using this technique, we developed methods for noncompetitive and competitive immunoassays, using thyroid stimulating hormone (TSH) and 17ß-estradiol (E2) as model analytes, respectively. We show that, compared to conventional methods, the new DMF approach reported here reduced reagent volumes and analysis time by 100-fold and 10-fold, respectively, while retaining a level of analytical performance required for clinical screening. Thus, we propose that the new technique has great potential for eventual use in a fast, low-waste, and inexpensive instrument for the quantitative analysis of proteins and small molecules in low sample volumes.


Subject(s)
Estradiol/analysis , Immunoassay/instrumentation , Magnetics/instrumentation , Microfluidic Analytical Techniques/instrumentation , Thyrotropin/analysis , Equipment Design , Humans , Immunoassay/economics , Limit of Detection , Magnetics/economics , Microfluidic Analytical Techniques/economics , Sample Size , Time Factors
13.
Anal Bioanal Chem ; 399(1): 337-45, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21057776

ABSTRACT

A digital microfluidic (DMF) device was applied to a heterogeneous sandwich immunoassay. The digital approach to microfluidics manipulates samples and reagents in the form of discrete droplets, as opposed to the streams of fluid used in microchannels. Since droplets are manipulated on relatively generic 2-D arrays of electrodes, DMF devices are straightforward to use, and are reconfigurable for any desired combination of droplet operations. This flexibility makes them suitable for a wide range of applications, especially those requiring long, multistep protocols such as immunoassays. Here, we developed an immunoassay on a DMF device using Human IgG as a model analyte. To capture the analyte, an anti-IgG antibody was physisorbed on the hydrophobic surface of a DMF device, and DMF actuation was used for all washing and incubation steps. The bound analyte was detected using FITC-labeled anti-IgG, and fluorescence after the final wash was measured in a fluorescence plate reader. A non-ionic polymer surfactant, Pluronic F-127, was added to sample and detection antibody solutions to control non-specific binding and aid in movement via DMF. Sample and reagent volumes were reduced by nearly three orders of magnitude relative to conventional multiwell plate methods. Since droplets are in constant motion, the antibody-antigen binding kinetics is not limited by diffusion, and total analysis times were reduced to less than 2.5 h per assay. A multiplexed device comprising several DMF platforms wired in series further increased the throughput of the technique. A dynamic range of approximately one order of magnitude was achieved, with reproducibility similar to the assay when performed in a 96-well plate. In bovine serum samples spiked with human IgG, the target molecule was successfully detected in the presence of a 100-fold excess of bovine IgG. It was concluded that the digital microfluidic format is capable of carrying out qualitative and quantitative sandwich immunoassays with a dramatic reduction in reagent usage and analysis time compared to macroscale methods.


Subject(s)
Immunoassay/methods , Immunoglobulin G/chemistry , Microfluidics/methods , Animals , Cattle , Humans , Immunoassay/instrumentation , Kinetics , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Microfluidics/instrumentation , Protein Binding , Serum/chemistry
14.
Nat Commun ; 12(1): 4031, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34188042

ABSTRACT

The response of patients with recurrent glioblastoma multiforme to neoadjuvant immune checkpoint blockade has been challenging to interpret due to the inter-patient and intra-tumor heterogeneity. We report on a comparative analysis of tumor tissues collected from patients with recurrent glioblastoma and high-risk melanoma, both treated with neoadjuvant checkpoint blockade. We develop a framework that uses multiplex spatial protein profiling, machine learning-based image analysis, and data-driven computational models to investigate the pathophysiological and molecular factors within the tumor microenvironment that influence treatment response. Using melanoma to guide the interpretation of glioblastoma analyses, we interrogate the protein expression in microscopic compartments of tumors, and determine the correlates of cytotoxic CD8+ T cells, tumor growth, treatment response, and immune cell-cell interaction. This work reveals similarities shared between glioblastoma and melanoma, immunosuppressive factors that are unique to the glioblastoma microenvironment, and potential co-targets for enhancing the efficacy of neoadjuvant immune checkpoint blockade.


Subject(s)
Brain Neoplasms/drug therapy , CTLA-4 Antigen/antagonists & inhibitors , Glioblastoma/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/analysis , Brain Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Female , Glioblastoma/pathology , Humans , Ipilimumab/therapeutic use , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Nivolumab/therapeutic use , Treatment Outcome , Tumor Microenvironment/immunology
16.
Anal Bioanal Chem ; 397(3): 991-1007, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20422163

ABSTRACT

Immunoassays have greatly benefited from miniaturization in microfluidic systems. This review, which summarizes developments in microfluidics-based immunoassays since 2000, includes four sections, focusing on the configurations of immunoassays that have been implemented in microfluidics, the main fluid handling modalities that have been used for microfluidic immunoassays, multiplexed immunoassays in microfluidic platforms, and the emergence of label-free detection techniques. The field of microfluidic immunoassays is continuously improving and has great promise for the future.


Subject(s)
Immunoassay/instrumentation , Microfluidic Analytical Techniques/instrumentation , Microfluidics/instrumentation , Animals , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Equipment Design , Humans , Immunoassay/methods , Microfluidic Analytical Techniques/methods , Microfluidics/methods
17.
Nat Commun ; 11(1): 4830, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32973134

ABSTRACT

Non-invasively probing metabolites within single live cells is highly desired but challenging. Here we utilize Raman spectro-microscopy for spatial mapping of metabolites within single cells, with the specific goal of identifying druggable metabolic susceptibilities from a series of patient-derived melanoma cell lines. Each cell line represents a different characteristic level of cancer cell de-differentiation. First, with Raman spectroscopy, followed by stimulated Raman scattering (SRS) microscopy and transcriptomics analysis, we identify the fatty acid synthesis pathway as a druggable susceptibility for differentiated melanocytic cells. We then utilize hyperspectral-SRS imaging of intracellular lipid droplets to identify a previously unknown susceptibility of lipid mono-unsaturation within de-differentiated mesenchymal cells with innate resistance to BRAF inhibition. Drugging this target leads to cellular apoptosis accompanied by the formation of phase-separated intracellular membrane domains. The integration of subcellular Raman spectro-microscopy with lipidomics and transcriptomics suggests possible lipid regulatory mechanisms underlying this pharmacological treatment. Our method should provide a general approach in spatially-resolved single cell metabolomics studies.


Subject(s)
Melanoma/metabolism , Metabolomics/methods , Microscopy/methods , Spectrum Analysis, Raman/methods , Apoptosis , Cell Line, Tumor , Fatty Acids/metabolism , Humans , Lipid Droplets , Lipid Metabolism , Lipidomics , Lipids , Oleic Acid , Stearoyl-CoA Desaturase/metabolism , Transcriptome
18.
Lab Chip ; 19(18): 3011-3021, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31502632

ABSTRACT

Adaptive immunity is based on peptide antigen recognition. Our ability to harness the immune system for therapeutic gain relies on the discovery of the T cell receptor (TCR) genes that selectively target antigens from infections, mutated proteins, and foreign agents. Here we present a method that selectively labels peptide antigen-specific CD8+ T cells using magnetic nanoparticles functionalized with peptide-MHC tetramers, isolates these specific cells within an integrated microfluidic device, and directly amplifies the TCR genes for sequencing. Critically, the identity of the peptide recognized by the TCR is preserved, providing the link between peptide and gene. The platform requires inputs on the order of just 100 000 CD8+ T cells, can be multiplexed for simultaneous analysis of multiple peptides, and performs sorting and isolation on chip. We demonstrate 1000-fold sensitivity enhancement of detecting antigen-specific TCRs relative to bulk analysis and simultaneous capture of two virus antigen-specific TCRs from a population of T cells.


Subject(s)
Antigens/genetics , Microfluidic Analytical Techniques , Receptors, Antigen, T-Cell/genetics , CD8-Positive T-Lymphocytes , Cells, Cultured , Humans , Magnetite Nanoparticles/chemistry , Microfluidic Analytical Techniques/instrumentation , Particle Size , Reverse Transcriptase Polymerase Chain Reaction
19.
Cell Rep ; 28(10): 2728-2738.e7, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31484081

ABSTRACT

Neoantigen-specific T cells are increasingly viewed as important immunotherapy effectors, but physically isolating these rare cell populations is challenging. Here, we describe a sensitive method for the enumeration and isolation of neoantigen-specific CD8+ T cells from small samples of patient tumor or blood. The method relies on magnetic nanoparticles that present neoantigen-loaded major histocompatibility complex (MHC) tetramers at high avidity by barcoded DNA linkers. The magnetic particles provide a convenient handle to isolate the desired cell populations, and the barcoded DNA enables multiplexed analysis. The method exhibits superior recovery of antigen-specific T cell populations relative to literature approaches. We applied the method to profile neoantigen-specific T cell populations in the tumor and blood of patients with metastatic melanoma over the course of anti-PD1 checkpoint inhibitor therapy. We show that the method has value for monitoring clinical responses to cancer immunotherapy and might help guide the development of personalized mutational neoantigen-specific T cell therapies and cancer vaccines.


Subject(s)
Antigens, Neoplasm/blood , Melanoma/blood , Melanoma/immunology , T-Lymphocytes/immunology , Biopsy , HEK293 Cells , Humans , Immunotherapy , Jurkat Cells , Kinetics , Lymphocytes, Tumor-Infiltrating/immunology , Magnetite Nanoparticles/chemistry , Major Histocompatibility Complex , Melanoma/pathology , Melanoma/secondary , Nucleic Acids/metabolism , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/metabolism , Reproducibility of Results , Tomography, X-Ray Computed
20.
Sci Transl Med ; 10(438)2018 04 25.
Article in English | MEDLINE | ID: mdl-29695457

ABSTRACT

Serosurveys are useful for assessing population susceptibility to vaccine-preventable disease outbreaks. Although at-risk populations in remote areas could benefit from this type of information, they face several logistical barriers to implementation, such as lack of access to centralized laboratories, cold storage, and transport of samples. We describe a potential solution: a compact and portable, field-deployable, point-of-care system relying on digital microfluidics that can rapidly test a small volume of capillary blood for disease-specific antibodies. This system uses inexpensive, inkjet-printed digital microfluidic cartridges together with an integrated instrument to perform enzyme-linked immunosorbent assays (ELISAs). We performed a field validation of the system's analytical performance at Kakuma refugee camp, a remote setting in northwestern Kenya, where we tested children aged 9 to 59 months and caregivers for measles and rubella immunoglobulin G (IgG). The IgG assays were determined to have sensitivities of 86% [95% confidence interval (CI), 79 to 91% (measles)] and 81% [95% CI, 73 to 88% (rubella)] and specificities of 80% [95% CI, 49 to 94% (measles)] and 91% [95% CI, 76 to 97% (rubella)] (measles, n = 140; rubella, n = 135) compared with reference tests (measles IgG and rubella IgG ELISAs from Siemens Enzygnost) conducted in a centralized laboratory. These results demonstrate a potential role for this point-of-care system in global serological surveillance, particularly in remote areas with limited access to centralized laboratories.


Subject(s)
Immunoassay/methods , Microfluidics/methods , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Infant , Male , Point-of-Care Systems
SELECTION OF CITATIONS
SEARCH DETAIL