Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 41
1.
Sci Signal ; 16(813): eadl3956, 2023 11 28.
Article En | MEDLINE | ID: mdl-38015912

Programmed cell death molecule 1 (PD-1) is a negative regulator of T cell activation; however, the mechanisms by which it acts are unclear. In this issue of Science Signaling, Paillon et al. show that PD-1 inhibits actin cytoskeletal rearrangements and associated effector responses in cytotoxic T cells.


Actins , Programmed Cell Death 1 Receptor , Actins/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Immunological Synapses , Actin Cytoskeleton/metabolism , Cytoskeleton , Lymphocyte Activation
2.
bioRxiv ; 2023 Oct 04.
Article En | MEDLINE | ID: mdl-37873483

Cytotoxic T lymphocytes (CTLs) carry out immunosurveillance by scanning target cells of diverse physical properties for the presence of antigens. While the recognition of cognate antigen by the T cell receptor is the primary signal for CTL activation, it has become increasingly clear that the mechanical stiffness of target cells plays an important role in antigen-triggered T cell responses. However, the molecular machinery within CTLs that transduces the mechanical information of tumor cells remains unclear. We find that CTL's mechanosensitive ability requires the activity of the actin-organizing protein Wiskott-Aldrich Syndrome Protein (WASP). WASP activation is modulated by the mechanical properties of antigen-presenting contexts across a wide range of target cell stiffnesses and activated WASP then mediates mechanosensitive activation of early TCR signaling markers in the CTL. Our results provide a molecular link between antigen mechanosensing and CTL immune response and suggest that CTL-intrinsic cytoskeletal organizing principles enable the processing of mechanical information from diverse target cells.

3.
Front Immunol ; 14: 1292166, 2023.
Article En | MEDLINE | ID: mdl-38264664

Ovarian cancer (OC) is the third most common gynecological cancer and alone has an emergence rate of approximately 308,069 cases worldwide (2020) with dire survival rates. To put it into perspective, the mortality rate of OC is three times higher than that of breast cancer and it is predicted to only increase significantly by 2040. The primary reasons for such a high rate are that the physical symptoms of OC are detectable only during the advanced phase of the disease when resistance to chemotherapies is high and around 80% of the patients that do indeed respond to chemotherapy initially, show a poor prognosis subsequently. This highlights a pressing need to develop new and effective therapies to tackle advanced OC to improve prognosis and patient survival. A major advance in this direction is the emergence of combination immunotherapeutic methods to boost CD8+ T cell function to tackle OC. In this perspective, we discuss our view of the current state of some of the combination immunotherapies in the treatment of advanced OC, their limitations, and potential approaches toward a safer and more effective response.


Breast Neoplasms , Ovarian Neoplasms , Humans , Female , Immunotherapy , Carcinoma, Ovarian Epithelial , CD8-Positive T-Lymphocytes
5.
Int J Biol Macromol ; 169: 95-102, 2021 Feb 01.
Article En | MEDLINE | ID: mdl-33338527

Detection of metal catalyzed carbonylation in proteins is traditionally based on derivatization followed by detection and quantification via spectroscopy or immunodetection. However, these measure only cumulative carbonylation and do not identify the specific sites of modification within the protein. Recently, fluorescein thiosemicarbazide (FTC) based semi-microplate method was adapted for high throughput monitoring of carbonyl content during mAb process development, using size-exclusion chromatography followed by ultraviolet and fluorescence detection. Here, we have examined carbonylation in originators and 4 biosimilars of an IgG1 therapeutic monoclonal antibody, trastuzumab, a first line of therapy for HER2 positive breast cancer. The hyphenated RP-ESI-MS/MS detection was able to identify the location of each of the carbonylated amino acids for all products. The result is a comprehensive map of a total of 27 unique identified carbonylation sites of trastuzumab found across multiple batches of originator as well as marketed biosimilars. Our results demonstrate that although the different carbonylation sites are spread across different domains throughout the mAb sequence, the complementarity determining regions (CDRs) are free of carbonylation and all identified sites lie within the framework region of the variable domain. Moreover, the constant- heavy domain 3 (CH3) region seems to be particularly resistant to process induced carbonylation.


Biosimilar Pharmaceuticals/chemistry , Protein Carbonylation/physiology , Trastuzumab/chemistry , Amino Acid Sequence/genetics , Amino Acids/genetics , Antibodies, Monoclonal/chemistry , Chromatography, Gel/methods , Fluoresceins/chemistry , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Tandem Mass Spectrometry/methods
7.
Sci Rep ; 10(1): 3317, 2020 Feb 19.
Article En | MEDLINE | ID: mdl-32076101

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
EMBO J ; 39(5): e102783, 2020 03 02.
Article En | MEDLINE | ID: mdl-31894880

When migratory T cells encounter antigen-presenting cells (APCs), they arrest and form radially symmetric, stable intercellular junctions termed immunological synapses which facilitate exchange of crucial biochemical information and are critical for T-cell immunity. While the cellular processes underlying synapse formation have been well characterized, those that maintain the symmetry, and thereby the stability of the synapse, remain unknown. Here we identify an antigen-triggered mechanism that actively promotes T-cell synapse symmetry by generating cytoskeletal tension in the plane of the synapse through focal nucleation of actin via Wiskott-Aldrich syndrome protein (WASP), and contraction of the resultant actin filaments by myosin II. Following T-cell activation, WASP is degraded, leading to cytoskeletal unraveling and tension decay, which result in synapse breaking. Thus, our study identifies and characterizes a mechanical program within otherwise highly motile T cells that sustains the symmetry and stability of the T cell-APC synaptic contact.


Antigen-Presenting Cells/metabolism , Immunological Synapses/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Antigen-Presenting Cells/immunology , Cell Movement , Cytoskeleton/metabolism , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein/genetics
9.
Oman J Ophthalmol ; 13(3): 149-151, 2020.
Article En | MEDLINE | ID: mdl-33542604

Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder. Here, we present a rare case of a middle-aged male, diagnosed with SLE, manifesting as bilateral central retinal artery occlusion (CRAO). Severe ocular complications such as CRAO may occur during an acute flare or even early in the disease process. It is important to recognize this potentially devastating complication.

10.
J Immunol ; 204(1): 49-57, 2020 01 01.
Article En | MEDLINE | ID: mdl-31740487

The control of cytoskeletal dynamics by dedicator of cytokinesis 2 (DOCK2), a hematopoietic cell-specific actin effector protein, has been implicated in TCR signaling and T cell migration. Biallelic mutations in Dock2 have been identified in patients with a recessive form of combined immunodeficiency with defects in T, B, and NK cell activation. Surprisingly, we show in this study that certain immune functions of CD8+ T cells are enhanced in the absence of DOCK2. Dock2-deficient mice have a pronounced expansion of their memory T cell compartment. Bone marrow chimera and adoptive transfer studies indicate that these memory T cells develop in a cell-intrinsic manner following thymic egress. Transcriptional profiling, TCR repertoire analyses, and cell surface marker expression indicate that Dock2-deficient naive CD8+ T cells directly convert into virtual memory cells without clonal effector T cell expansion. This direct conversion to memory is associated with a selective increase in TCR sensitivity to self-peptide MHC in vivo and an enhanced response to weak agonist peptides ex vivo. In contrast, the response to strong agonist peptides remains unaltered in Dock2-deficient T cells. Collectively, these findings suggest that the regulation of the actin dynamics by DOCK2 enhances the threshold for entry into the virtual memory compartment by negatively regulating tonic TCR triggering in response to weak agonists.


CD8-Positive T-Lymphocytes/immunology , GTPase-Activating Proteins/immunology , Guanine Nucleotide Exchange Factors/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Homeodomain Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
11.
Trends Immunol ; 40(11): 977-980, 2019 11.
Article En | MEDLINE | ID: mdl-31645298

T cells comprise functionally diverse subtypes. Although activated via a conserved scheme of antigen recognition by their T cell receptor, they elicit heterogeneous activation and effector responses. Such functional diversity has been appreciated in gene expression studies, functional assays, and disease models. Yet, our understanding of the principles underlying T cell subtype-specific activation and antigen recognition in the immunological synapse remains limited. This is primarily due to difficulties in primary T cell visualization at high spatiotemporal resolution and the adoption of tractable transformed T cell systems for cell biological experiments that may not correctly represent primary T cell constitutional diversity. Here, we discuss recent findings regarding the architectural and dynamic diversity of the immunological synapse and state-of-the-art methodologies that can be utilized to provide clues on how biological and biophysical differences in synaptic make-up could govern functional divergences in T cell subtypes.


B-Lymphocytes/immunology , Dendritic Cells/immunology , Immunological Synapses/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Humans , Lymphocyte Activation , Receptor Cross-Talk , Signal Transduction
12.
J Cell Sci ; 133(5)2019 09 04.
Article En | MEDLINE | ID: mdl-31413071

Cytoskeletal actin dynamics are crucial for the activation of T-cells. Immortalised Jurkat T-cells have been the model system of choice to examine and correlate the dynamics of the actin cytoskeleton and the immunological synapse leading to T-cell activation. However, it has remained unclear whether immortalised cellular systems, such as Jurkat T-cells can recapitulate the cytoskeletal behaviour of primary T-cells. Studies delineating the cytoskeletal behaviour of Jurkat T-cells in comparison to primary T-cells are lacking. Here, we employ live-cell super-resolution microscopy to investigate the cytoskeletal actin organisation and dynamics of living primary and immortalised Jurkat T-cells at the appropriate spatiotemporal resolution. Under comparable activation conditions, we found differences in the architectural organisation and dynamics of Jurkat and primary mouse and human T-cells. Although the three main actin network architectures in Jurkat T-cells were reminiscent of primary T-cells, there were differences in the organisation and molecular mechanisms underlying these networks. Our results highlight mechanistic distinctions in the T-cell model system most utilised to study cytoskeletal actin dynamics.


Actin Cytoskeleton/metabolism , Actins/metabolism , Immunological Synapses/metabolism , T-Lymphocytes/cytology , Animals , Gene Rearrangement, T-Lymphocyte , Humans , Jurkat Cells , Lymphocyte Activation , Mice , Models, Biological , Receptors, Antigen, T-Cell/genetics , Signal Transduction
13.
Science ; 365(6449): 162-168, 2019 07 12.
Article En | MEDLINE | ID: mdl-31296767

Chimeric antigen receptor-T cell (CAR-T) therapy has been effective in the treatment of hematologic malignancies, but it has shown limited efficacy against solid tumors. Here we demonstrate an approach to enhancing CAR-T function in solid tumors by directly vaccine-boosting donor cells through their chimeric receptor in vivo. We designed amphiphile CAR-T ligands (amph-ligands) that, upon injection, trafficked to lymph nodes and decorated the surfaces of antigen-presenting cells, thereby priming CAR-Ts in the native lymph node microenvironment. Amph-ligand boosting triggered massive CAR-T expansion, increased donor cell polyfunctionality, and enhanced antitumor efficacy in multiple immunocompetent mouse tumor models. We demonstrate two approaches to generalizing this strategy to any chimeric antigen receptor, enabling this simple non-human leukocyte antigen-restricted approach to enhanced CAR-T functionality to be applied to existing CAR-T designs.


Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , HEK293 Cells , Humans , Immunization, Secondary , K562 Cells , Mice
14.
Cell Rep ; 26(12): 3369-3379.e5, 2019 03 19.
Article En | MEDLINE | ID: mdl-30893608

Cytoskeletal actin dynamics is essential for T cell activation. Here, we show evidence that the binding kinetics of the antigen engaging the T cell receptor influences the nanoscale actin organization and mechanics of the immune synapse. Using an engineered T cell system expressing a specific T cell receptor and stimulated by a range of antigens, we found that the peak force experienced by the T cell receptor during activation was independent of the unbinding kinetics of the stimulating antigen. Conversely, quantification of the actin retrograde flow velocity at the synapse revealed a striking dependence on the antigen unbinding kinetics. These findings suggest that the dynamics of the actin cytoskeleton actively adjusted to normalize the force experienced by the T cell receptor in an antigen-specific manner. Consequently, tuning actin dynamics in response to antigen kinetics may thus be a mechanism that allows T cells to adjust the lengthscale and timescale of T cell receptor signaling.


Actin Cytoskeleton/immunology , Lymphocyte Activation , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Humans , Jurkat Cells , T-Lymphocytes/cytology
15.
Science ; 363(6427): 649-654, 2019 02 08.
Article En | MEDLINE | ID: mdl-30573546

In vaccine design, antigens are often arrayed in a multivalent nanoparticle form, but in vivo mechanisms underlying the enhanced immunity elicited by such vaccines remain poorly understood. We compared the fates of two different heavily glycosylated HIV antigens, a gp120-derived mini-protein and a large, stabilized envelope trimer, in protein nanoparticle or "free" forms after primary immunization. Unlike monomeric antigens, nanoparticles were rapidly shuttled to the follicular dendritic cell (FDC) network and then concentrated in germinal centers in a complement-, mannose-binding lectin (MBL)-, and immunogen glycan-dependent manner. Loss of FDC localization in MBL-deficient mice or via immunogen deglycosylation significantly affected antibody responses. These findings identify an innate immune-mediated recognition pathway promoting antibody responses to particulate antigens, with broad implications for humoral immunity and vaccine design.


AIDS Vaccines/immunology , Antibody Formation , Germinal Center/immunology , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , Immunity, Innate , Polysaccharides/immunology , Animals , Antibodies, Neutralizing/immunology , Complement Fixation Tests , Complement System Proteins/immunology , Dendritic Cells/immunology , Female , HIV Antibodies/immunology , HIV Infections/prevention & control , Liposomes , Mannose-Binding Lectin/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Multiprotein Complexes , Nanoparticles , Receptors, Complement/immunology
16.
J Clin Invest ; 128(10): 4604-4621, 2018 10 01.
Article En | MEDLINE | ID: mdl-30106752

Regulatory T cells (Tregs) are critical for maintaining immune homeostasis. However, current Treg immunotherapies do not optimally treat inflammatory diseases in patients. Understanding the cellular processes that control Treg function may allow for the augmentation of therapeutic efficacy. In contrast to activated conventional T cells, in which protein kinase C-θ (PKC-θ) localizes to the contact point between T cells and antigen-presenting cells, in human and mouse Tregs, PKC-θ localizes to the opposite end of the cell in the distal pole complex (DPC). Here, using a phosphoproteomic screen, we identified the intermediate filament vimentin as a PKC-θ phospho target and show that vimentin forms a DPC superstructure on which PKC-θ accumulates. Treatment of mouse Tregs with either a clinically relevant PKC-θ inhibitor or vimentin siRNA disrupted vimentin and enhanced Treg metabolic and suppressive activity. Moreover, vimentin-disrupted mouse Tregs were significantly better than controls at suppressing alloreactive T cell priming in graft-versus-host disease (GVHD) and GVHD lethality, using a complete MHC-mismatch mouse model of acute GVHD (C57BL/6 donor into BALB/c host). Interestingly, vimentin disruption augmented the suppressor function of PKC-θ-deficient mouse Tregs. This suggests that enhanced Treg activity after PKC-θ inhibition is secondary to effects on vimentin, not just PKC-θ kinase activity inhibition. Our data demonstrate that vimentin is a key metabolic and functional controller of Treg activity and provide proof of principle that disruption of vimentin is a feasible, translationally relevant method to enhance Treg potency.


Antigen-Presenting Cells/immunology , Graft vs Host Disease/immunology , Intermediate Filaments/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Vimentin/immunology , Animals , Antigen-Presenting Cells/pathology , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Humans , Intermediate Filaments/genetics , Intermediate Filaments/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Protein Kinase C-theta/genetics , Protein Kinase C-theta/immunology , T-Lymphocytes, Regulatory/pathology , Vimentin/genetics
17.
JCI Insight ; 2(19)2017 10 05.
Article En | MEDLINE | ID: mdl-28978806

Patients deficient in the guanine nucleotide exchange factor DOCK8 have decreased numbers and impaired in vitro function of Tregs and make autoantibodies, but they seldom develop autoimmunity. We show that, similarly, Dock8-/- mice have decreased numbers and impaired in vitro function of Tregs but do not develop autoimmunity. In contrast, mice with selective DOCK8 deficiency in Tregs develop lymphoproliferation, autoantibodies, and gastrointestinal inflammation, despite a normal percentage and in vitro function of Tregs, suggesting that deficient T effector cell function might protect DOCK8-deficient patients from autoimmunity. We demonstrate that DOCK8 associates with STAT5 and is important for IL-2-driven STAT5 phosphorylation in Tregs. DOCK8 localizes within the lamellar actin ring of the Treg immune synapse (IS). Dock8-/- Tregs have abnormal TCR-driven actin dynamics, decreased adhesiveness, an altered gene expression profile, an unstable IS with decreased recruitment of signaling molecules, and impaired transendocytosis of the costimulatory molecule CD86. These data suggest that DOCK8 enforces immunological tolerance by promoting IL-2 signaling, TCR-driven actin dynamics, and the IS in Tregs.


Guanine Nucleotide Exchange Factors/immunology , Immune Tolerance/immunology , Immunological Synapses/immunology , Interleukin-2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoantibodies/biosynthesis , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Gastroenteritis/immunology , Guanine Nucleotide Exchange Factors/deficiency , Inflammation/immunology , Lymph Nodes/immunology , Mice, Knockout , Phosphorylation/immunology , STAT5 Transcription Factor/metabolism , Weight Gain/immunology
18.
Sci Rep ; 7(1): 4258, 2017 06 26.
Article En | MEDLINE | ID: mdl-28652577

In vitro induced human regulatory T cells (iTregs) have demonstrated in vivo therapeutic utility, but pathways regulating their function have not been elucidated. Here, we report that human iTregs generated in vitro from naïve cord blood cells preferentially recruit Disc large homolog 1 (Dlgh1) and exclude protein kinase C (PKC)-θ from immunological synapses formed on supported lipid bilayers with laterally mobile ICAM-1 and anti-CD3 mAb. Also, iTregs display elevated Dlgh1 overall and Dlgh1-dependent p38 phosphorylation, higher levels of phosphatase and tensin homolog (PTEN), and diminished Akt phosphorylation. Pharmacological interruption of PKC-θ increases and Dlgh1 silencing decreases the ability of iTregs to suppress interferon-γ production by CD4+CD25- effector T cells (Teff). Comparison with expanded cord blood-derived CD4+CD25hi tTreg and expanded Teffs from the same donors indicate that iTreg are intermediate between expanded CD4+CD25hi tTregs and Teffs, whereas modulation of suppressive activities by PKC-θ and Dlgh1 signaling pathways are shared.


Adaptor Proteins, Signal Transducing/genetics , Immunological Synapses/genetics , Membrane Proteins/genetics , Protein Kinase C-theta/genetics , T-Lymphocytes, Regulatory/metabolism , Adaptor Proteins, Signal Transducing/immunology , CD4 Antigens/genetics , Cell Differentiation/genetics , Discs Large Homolog 1 Protein , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , Immunological Synapses/metabolism , Intercellular Adhesion Molecule-1/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Lipid Bilayers/immunology , Lipid Bilayers/metabolism , Lymphocyte Activation , Membrane Proteins/immunology , Phosphorylation , Protein Kinase C-theta/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , p38 Mitogen-Activated Protein Kinases/genetics
19.
ACS Nano ; 11(3): 3089-3100, 2017 03 28.
Article En | MEDLINE | ID: mdl-28231431

Adoptive cell therapy (ACT) has achieved striking efficacy in B-cell leukemias, but less success treating other cancers, in part due to the rapid loss of ACT T-cell effector function in vivo due to immunosuppression in solid tumors. Transforming growth factor-ß (TGF-ß) signaling is an important mechanism of immune suppression in the tumor microenvironment, but systemic inhibition of TGF-ß is toxic. Here we evaluated the potential of targeting a small molecule inhibitor of TGF-ß to ACT T-cells using PEGylated immunoliposomes. Liposomes were prepared that released TGF-ß inhibitor over ∼3 days in vitro. We compared the impact of targeting these drug-loaded vesicles to T-cells via an internalizing receptor (CD90) or noninternalizing receptor (CD45). When lymphocytes were preloaded with immunoliposomes in vitro prior to adoptive therapy, vesicles targeted to both CD45 and CD90 promoted enhanced T-cell expression of granzymes relative to free systemic drug administration, but only targeting to CD45 enhanced accumulation of granzyme-expressing T-cells in tumors, which correlated with the greatest enhancement of T-cell antitumor activity. By contrast, when administered i.v. to target T-cells in vivo, only targeting of a CD90 isoform expressed exclusively by the donor T-cells led to greater tumor regression over equivalent doses of free systemic drug. These results suggest that in vivo, targeting of receptors uniquely expressed by donor T-cells is of paramount importance for maximal efficacy. This immunoliposome strategy should be broadly applicable to target exogenous or endogenous T-cells and defines parameters to optimize delivery of supporting (or suppressive) drugs to these important immune effectors.


Drug Delivery Systems , Immunologic Factors/administration & dosage , Immunologic Factors/immunology , Immunotherapy, Adoptive , Melanoma, Experimental/therapy , Small Molecule Libraries/pharmacology , T-Lymphocytes/immunology , Animals , Female , Immunologic Factors/chemistry , Leukocyte Common Antigens/antagonists & inhibitors , Leukocyte Common Antigens/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Small Molecule Libraries/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/transplantation , Thy-1 Antigens/antagonists & inhibitors , Thy-1 Antigens/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
20.
Biomaterials ; 117: 44-53, 2017 02.
Article En | MEDLINE | ID: mdl-27936416

Cytotoxic T-Lymphocytes (CTLs) kill pathogen-infected or transformed cells following interaction of their T-cell receptors (TCRs) with foreign (e.g. virus-derived) peptides bound to MHC-I molecules on the target cell. TCR binding triggers CTLs to secrete perforin, which forms pores in the target cell membrane, promoting target death. Here, we show that by conjugating drug-loaded lipid nanoparticles to the surface of CTLs, their lytic machinery can be co-opted to lyse the cell-bound drug carrier, providing triggered release of drug cargo upon target cell recognition. Protein encapsulated in T-cell-bound nanoparticles was released following culture of CTLs with target cells in an antigen dose- and perforin-dependent manner and coincided with target cell lysis. Using this approach, we demonstrate the capacity of HIV-specific CTLs to deliver an immunotherapeutic agent to an anatomical site of viral replication. This strategy provides a novel means to couple drug delivery to the action of therapeutic cells in vivo.


Anti-HIV Agents/administration & dosage , Autoantigens/immunology , HIV Infections/immunology , HIV Infections/therapy , Nanocapsules/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , Animals , Delayed-Action Preparations/administration & dosage , Mice
...