Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Adv Mater ; 36(23): e2310043, 2024 Jun.
Article En | MEDLINE | ID: mdl-38358310

T cells are critical mediators of antigen-specific immune responses and are common targets for immunotherapy. Biomaterial scaffolds have previously been used to stimulate antigen-presenting cells to elicit antigen-specific immune responses; however, structural and molecular features that directly stimulate and expand naïve, endogenous, tumor-specific T cells in vivo have not been defined. Here, an artificial lymph node (aLN) matrix is created, which consists of an extracellular matrix hydrogel conjugated with peptide-loaded-MHC complex (Signal 1), the co-stimulatory signal anti-CD28 (Signal 2), and a tethered IL-2 (Signal 3), that can bypass challenges faced by other approaches to activate T cells in situ such as vaccines. This dynamic immune-stimulating platform enables direct, in vivo antigen-specific CD8+ T cell stimulation, as well as recruitment and coordination of host immune cells, providing an immuno-stimulatory microenvironment for antigen-specific T cell activation and expansion. Co-injecting the aLN with naïve, wild-type CD8+ T cells results in robust activation and expansion of tumor-targeted T cells that kill target cells and slow tumor growth in several distal tumor models. The aLN platform induces potent in vivo antigen-specific CD8+ T cell stimulation without the need for ex vivo priming or expansion and enables in situ manipulation of antigen-specific responses for immunotherapies.


CD8-Positive T-Lymphocytes , Lymph Nodes , Animals , Lymph Nodes/immunology , CD8-Positive T-Lymphocytes/immunology , Mice , Lymphocyte Activation , Hydrogels/chemistry , Immunotherapy/methods , Extracellular Matrix/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Humans , Interleukin-2/metabolism , Peptides/chemistry , Cell Line, Tumor , Mice, Inbred C57BL
2.
JCI Insight ; 7(18)2022 09 22.
Article En | MEDLINE | ID: mdl-36134660

Cross-reactive immunity between SARS-CoV-2 and other related coronaviruses has been well-documented, and it may play a role in preventing severe COVID-19. Epidemiological studies early in the pandemic showed a geographical association between high influenza vaccination rates and lower incidence of SARS-CoV-2 infection. We, therefore, analyzed whether exposure to influenza A virus (IAV) antigens could influence the T cell repertoire in response to SARS-CoV-2, indicating a heterologous immune response between these 2 unrelated viruses. Using artificial antigen-presenting cells (aAPCs) combined with real-time reverse-transcription PCR (RT-qPCR), we developed a sensitive assay to quickly screen for antigen-specific T cell responses and detected a significant correlation between responses to SARS-CoV-2 epitopes and IAV dominant epitope (M158-66). Further analysis showed that some COVID-19 convalescent donors exhibited both T cell receptor (TCR) specificity and functional cytokine responses to multiple SARS-CoV-2 epitopes and M158-66. Utilizing an aAPC-based stimulation/expansion assay, we detected cross-reactive T cells with specificity to SARS-CoV-2 and IAV. In addition, TCR sequencing of the cross-reactive and IAV-specific T cells revealed similarities between the TCR repertoires of the two populations. These results indicate that heterologous immunity shaped by our exposure to other unrelated endemic viruses may affect our immune response to novel viruses such as SARS-CoV-2.


COVID-19 , Influenza, Human , Antigens, Viral , CD8-Positive T-Lymphocytes , Cytokines , Epitopes , Humans , Receptors, Antigen, T-Cell , SARS-CoV-2
3.
Nano Lett ; 20(9): 6289-6298, 2020 09 09.
Article En | MEDLINE | ID: mdl-32594746

T cells are critical players in disease; yet, their antigen-specificity has been difficult to identify, as current techniques are limited in terms of sensitivity, throughput, or ease of use. To address these challenges, we increased the throughput and translatability of magnetic nanoparticle-based artificial antigen presenting cells (aAPCs) to enrich and expand (E+E) murine or human antigen-specific T cells. We streamlined enrichment, expansion, and aAPC production processes by enriching CD8+ T cells directly from unpurified immune cells, increasing parallel processing capacity of aAPCs in a 96-well plate format, and designing an adaptive aAPC that enables multiplexed aAPC construction for E+E and detection. We applied these adaptive platforms to process and detect CD8+ T cells specific for rare cancer neoantigens, commensal bacterial cross-reactive epitopes, and human viral and melanoma antigens. These innovations dramatically increase the multiplexing ability and decrease the barrier to adopt for investigating antigen-specific T cell responses.


Nanoparticles , Neoplasms , Animals , Antigen-Presenting Cells , CD8-Positive T-Lymphocytes , Epitopes , Humans , Mice
4.
ACS Nano ; 9(7): 6861-71, 2015 Jul 28.
Article En | MEDLINE | ID: mdl-26171764

Adoptive immunotherapy (AIT) can mediate durable regression of cancer, but widespread adoption of AIT is limited by the cost and complexity of generating tumor-specific T cells. Here we develop an Enrichment + Expansion strategy using paramagnetic, nanoscale artificial antigen presenting cells (aAPC) to rapidly expand tumor-specific T cells from rare naïve precursors and predicted neo-epitope responses. Nano-aAPC are capable of enriching rare tumor-specific T cells in a magnetic column and subsequently activating them to induce proliferation. Enrichment + Expansion resulted in greater than 1000-fold expansion of both mouse and human tumor-specific T cells in 1 week, with nano-aAPC based enrichment conferring a proliferation advantage during both in vitro culture and after adoptive transfer in vivo. Robust T cell responses were seen not only for shared tumor antigens, but also for computationally predicted neo-epitopes. Streamlining the rapid generation of large numbers of tumor-specific T cells in a cost-effective fashion through Enrichment + Expansion can be a powerful tool for immunotherapy.


Antigen-Presenting Cells/cytology , Antigens, Neoplasm/immunology , Cell Separation/methods , Nanoparticles/chemistry , Adaptive Immunity , Animals , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/chemistry , Cell Line, Tumor , Cells, Cultured , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL
5.
ACS Nano ; 8(3): 2252-60, 2014 Mar 25.
Article En | MEDLINE | ID: mdl-24564881

Iron-dextran nanoparticles functionalized with T cell activating proteins have been used to study T cell receptor (TCR) signaling. However, nanoparticle triggering of membrane receptors is poorly understood and may be sensitive to physiologically regulated changes in TCR clustering that occur after T cell activation. Nano-aAPC bound 2-fold more TCR on activated T cells, which have clustered TCR, than on naive T cells, resulting in a lower threshold for activation. To enhance T cell activation, a magnetic field was used to drive aggregation of paramagnetic nano-aAPC, resulting in a doubling of TCR cluster size and increased T cell expansion in vitro and after adoptive transfer in vivo. T cells activated by nano-aAPC in a magnetic field inhibited growth of B16 melanoma, showing that this novel approach, using magnetic field-enhanced nano-aAPC stimulation, can generate large numbers of activated antigen-specific T cells and has clinically relevant applications for adoptive immunotherapy.


Lymphocyte Activation/drug effects , Magnetic Fields , Melanoma, Experimental/therapy , Nanoparticles , Receptors, Antigen, T-Cell/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , CD3 Complex/metabolism , Cell Proliferation/drug effects , Dextrans/chemistry , Immunotherapy, Adoptive , Iron/chemistry , Iron/pharmacology , Melanoma, Experimental/immunology , Mice , Protein Multimerization/drug effects , Protein Structure, Quaternary , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
6.
Nanomedicine ; 10(1): 119-29, 2014 Jan.
Article En | MEDLINE | ID: mdl-23891987

Artificial antigen presenting cells (aAPC), which deliver stimulatory signals to cytotoxic lymphocytes, are a powerful tool for both adoptive and active immunotherapy. Thus far, aAPC have been synthesized by coupling T cell activating proteins such as CD3 or MHC-peptide to micron-sized beads. Nanoscale platforms have different trafficking and biophysical interaction properties and may allow development of new immunotherapeutic strategies. We therefore manufactured aAPC based on two types of nanoscale particle platforms: biocompatible iron-dextran paramagnetic particles (50-100 nm in diameter) and avidin-coated quantum dot nanocrystals (~30 nm). Nanoscale aAPC induced antigen-specific T cell proliferation from mouse splenocytes and human peripheral blood T cells. When injected in vivo, both iron-dextran particles and quantum dot nanocrystals enhanced tumor rejection in a subcutaneous mouse melanoma model. This is the first description of nanoscale aAPC that induce antigen-specific T cell proliferation in vitro and lead to effective T cell stimulation and inhibition of tumor growth in vivo. FROM THE CLINICAL EDITOR: Artifical antigen presenting cells could revolutionize the field of cancer-directed immunotherapy. This team of investigators have manufactured two types of nanoscale particle platform-based aAPCs and demonstrates that both iron-dextran particles and quantum dot nanocrystals enhance tumor rejection in a melanoma model, providing the first description of nanoscale aAPCs that lead to effective T cell stimulation and inhibition of tumor growth.


Immunotherapy , Iron-Dextran Complex/therapeutic use , Melanoma/therapy , Nanoparticles/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Cell Proliferation/drug effects , Humans , Iron-Dextran Complex/immunology , Melanoma/immunology , Melanoma/pathology , Mice , Nanoparticles/therapeutic use , Quantum Dots/administration & dosage , Quantum Dots/chemistry
7.
Biophys J ; 103(9): 1890-8, 2012 Nov 07.
Article En | MEDLINE | ID: mdl-23199917

The structure of a T cell receptor (TCR) and its affinity for cognate antigen are fixed, but T cells regulate binding sensitivity through changes in lateral membrane organization. TCR microclusters formed upon antigen engagement participate in downstream signaling. Microclusters are also found 3-4 days after activation, leading to enhanced antigen binding upon rechallenge. However, others have found an almost complete loss of antigen binding four days after T cell activation, when TCR clusters are present. To resolve these contradictory results, we compared binding of soluble MHC-Ig dimers by transgenic T cells stimulated with a high (100 µM) or low (100 fM) dose of cognate antigen. Cells activated by a high dose of peptide bound sixfold lower amounts of CD8-dependent ligand K(b)-SIY than cells activated by a low dose of MHC/peptide. In contrast, both cell populations bound a CD8-independent ligand L(d)-QL9 equally well. Consistent with the differences between binding of CD8-dependent and CD8-independent peptide/MHC, Förster resonance energy transfer (FRET) measurements of molecular proximity reported little nanoscale association of TCR with CD8 (16 FRET units) compared to their association on cells stimulated by low antigen dose (62 FRET units). Loss of binding induced by changes in lateral organization of TCR and CD8 may serve as a regulatory mechanism to avoid excessive inflammation and immunopathology in response to aggressive infection.


CD8 Antigens/metabolism , Histocompatibility Antigens/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Fluorescence Resonance Energy Transfer , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Receptors, Antigen, T-Cell/genetics
8.
Biophys J ; 101(11): L57-9, 2011 Dec 07.
Article En | MEDLINE | ID: mdl-22261075

Changes in the clustering of surface receptors modulate cell responses to ligands. Hence, global measures of receptor clustering can be useful for characterizing cell states. Using T cell receptor for antigen as an example, we show that k-space image correlation spectroscopy of quantum dots blinking detects T cell receptor clusters on a scale of tens of nanometers and reports changes in clustering after T cell activation. Our results offer a general approach to the global analysis of lateral organization and receptor clustering in single cells, and can thus be applied when the cell type of interest is rare.


Nanoparticles/chemistry , Quantum Dots , Receptors, Antigen, T-Cell/immunology , Animals , Fluorescence , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Time Factors
...