Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
JCI Insight ; 6(20)2021 10 22.
Article En | MEDLINE | ID: mdl-34491911

The PD-1/PD-L1 pathway is a key immune checkpoint that regulates T cell activation. There is strong rationale to develop PD-1 agonists as therapeutics against autoimmunity, but progress in this area has been limited. Here, we generated T cell receptor (TCR) targeting, PD-1 agonist bispecifics called ImmTAAI molecules that mimic the ability of PD-L1 to facilitate the colocalization of PD-1 with the TCR complex at the target cell-T cell interface. PD-1 agonist ImmTAAI molecules specifically bound to target cells and were highly effective in activating the PD-1 receptor on interacting T cells to achieve immune suppression. Potent PD-1 antibody ImmTAAI molecules closely mimicked the mechanism of action of endogenously expressed PD-L1 in their localization to the target cell-T cell interface, inhibition of proximal TCR signaling events, and suppression of T cell function. At picomolar concentrations, these bispecifics suppressed cytokine production and inhibited CD8+ T cell-mediated cytotoxicity in vitro. Crucially, in soluble form, the PD-1 ImmTAAI molecules were inactive and, hence, could avoid systemic immunosuppression. This study outlines a promising new route to generate more effective, potent, tissue-targeted PD-1 agonists that can inhibit T cell function locally with the potential to treat autoimmune and chronic inflammatory diseases of high unmet need.


Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/antagonists & inhibitors , Humans
2.
Nanoscale ; 13(35): 15010-15020, 2021 Sep 17.
Article En | MEDLINE | ID: mdl-34533174

For effective targeted therapy of cancer with chemotherapy-loaded nanoparticles (NPs), antigens that are selective for cancer cells should be targeted to minimise off-tumour toxicity. Human leukocyte antigens (HLAs) are attractive cancer targets as they can present peptides from tumour-selective proteins on the cell surface, which can be recognised by T cells via T cell receptors (TCRs). In this study, docetaxel-loaded polymeric NPs were conjugated to recombinant affinity-enhanced TCRs to target breast cancer cells presenting a tumour-selective peptide-HLA complex. The TCR-conjugated nanoparticles enabled enhanced delivery of docetaxel and induced cell death through tumour-specific peptide-HLA targeting. These in vitro data demonstrate the potential of targeting tumour-restricted peptide-HLA epitopes using high affinity TCR-conjugated nanoparticles, representing a novel treatment strategy to deliver therapeutic drugs specifically to cancer cells.


Nanoparticles , Receptors, Antigen, T-Cell , Antigens, Neoplasm , Cell Line, Tumor , Docetaxel , Humans , T-Lymphocytes
3.
PLoS One ; 13(10): e0205491, 2018.
Article En | MEDLINE | ID: mdl-30321203

Robust preclinical testing is essential to predict clinical safety and efficacy and provide data to determine safe dose for first-in-man studies. There are a growing number of examples where the preclinical development of drugs failed to adequately predict clinical adverse events in part due to their assessment with inappropriate preclinical models. Preclinical investigations of T cell receptor (TCR)-based immunotherapies prove particularly challenging as these biologics are human-specific and thus the conventional testing in animal models is inadequate. As these molecules harness the full force of the immune system, and demonstrate tremendous potency, we set out to design a preclinical package that would ensure adequate evaluation of these therapeutics. Immune Mobilising Monoclonal TCR Against Cancer (ImmTAC) molecules are bi-specific biologics formed of an affinity-enhanced TCR fused to an anti-CD3 effector function. ImmTAC molecules are designed to activate human T lymphocytes and target peptides within the context of a human leukocyte antigen (HLA), thus require an intact human immune system and peptidome for suitable preclinical screening. Here we draw upon the preclinical testing of four ImmTAC molecules, including IMCgp100, the first ImmTAC molecule to reach the clinic, to present our comprehensive, informative and robust approach to in vitro preclinical efficacy and safety screening. This package comprises a broad range of cellular and molecular assays using human tissues and cultured cells to test efficacy, safety and specificity, and hence predict human responses in clinical trials. We propose that this entirely in vitro package offers a potential model to be applied to screening other TCR-based biologics.


Antibodies, Bispecific/pharmacology , Drug Screening Assays, Antitumor/methods , Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , In Vitro Techniques , Workflow
4.
Immunology ; 152(3): 425-438, 2017 11.
Article En | MEDLINE | ID: mdl-28640942

The success of immune system-based cancer therapies depends on a broad immune response engaging a range of effector cells and mechanisms. Immune mobilizing monoclonal T cell receptors (TCRs) against cancer (ImmTAC™ molecules: fusion proteins consisting of a soluble, affinity enhanced TCR and an anti-CD3 scFv antibody) were previously shown to redirect CD8+ and CD4+ T cells against tumours. Here we present evidence that IMCgp100 (ImmTAC recognizing a peptide derived from the melanoma-specific protein, gp100, presented by HLA-A*0201) efficiently redirects and activates effector and memory cells from both CD8+ and CD4+ repertoires. Using isolated subpopulations of T cells, we find that both terminally differentiated and effector memory CD8+ T cells redirected by IMCgp100 are potent killers of melanoma cells. Furthermore, CD4+ effector memory T cells elicit potent cytotoxic activity leading to melanoma cell killing upon redirection by IMCgp100. The majority of T cell subsets belonging to both the CD8+ and CD4+ repertoires secrete key pro-inflammatory cytokines (tumour necrosis factor-α, interferon-γ, interleukin-6) and chemokines (macrophage inflammatory protein-1α-ß, interferon-γ-inducible protein-10, monocyte chemoattractant protein-1). At an individual cell level, IMCgp100-redirected T cells display a polyfunctional phenotype, which is a hallmark of a potent anti-cancer response. This study demonstrates that IMCgp100 induces broad immune responses that extend beyond the induction of CD8+ T cell-mediated cytotoxicity. These findings are of particular importance because IMCgp100 is currently undergoing clinical trials as a single agent or in combination with check point inhibitors for patients with malignant melanoma.


CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Melanoma/therapy , Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Skin Neoplasms/therapy , gp100 Melanoma Antigen/immunology , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immunologic Memory/drug effects , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Phenotype , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors , gp100 Melanoma Antigen/metabolism
5.
Mol Ther ; 24(11): 1913-1925, 2016 Nov.
Article En | MEDLINE | ID: mdl-27401039

Persistence of human immunodeficiency virus (HIV) in a latent state in long-lived CD4+ T-cells is a major barrier to eradication. Latency-reversing agents that induce direct or immune-mediated cell death upon reactivation of HIV are a possible solution. However, clearance of reactivated cells may require immunotherapeutic agents that are fine-tuned to detect viral antigens when expressed at low levels. We tested the antiviral efficacy of immune-mobilizing monoclonal T-cell receptors against viruses (ImmTAVs), bispecific molecules that redirect CD8+ T-cells to kill HIV-infected CD4+ T-cells. T-cell receptors specific for an immunodominant Gag epitope, SL9, and its escape variants were engineered to achieve supraphysiological affinity and fused to a humanised CD3-specific single chain antibody fragment. Ex vivo polyclonal CD8+ T-cells were efficiently redirected by immune-mobilising monoclonal T-cell receptors against viruses to eliminate CD4+ T-cells from human histocompatibility leukocyte antigen (HLA)-A*0201-positive antiretroviral therapy-treated patients after reactivation of inducible HIV in vitro. The efficiency of infected cell elimination correlated with HIV Gag expression. Immune-mobilising monoclonal T-cell receptors against viruses have potential as a therapy to facilitate clearance of reactivated HIV reservoir cells.


HIV Antibodies/pharmacology , HIV Infections/drug therapy , HIV-1/physiology , Receptors, Antigen, T-Cell/immunology , Antibodies, Monoclonal/pharmacology , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/immunology , Humans , Virus Latency
6.
Sci Rep ; 6: 18851, 2016 Jan 13.
Article En | MEDLINE | ID: mdl-26758806

Natural T-cell responses generally lack the potency to eradicate cancer. Enhanced affinity T-cell receptors (TCRs) provide an ideal approach to target cancer cells, with emerging clinical data showing significant promise. Nevertheless, the risk of off target reactivity remains a key concern, as exemplified in a recent clinical report describing fatal cardiac toxicity, following administration of MAGE-A3 specific TCR-engineered T-cells, mediated through cross-reactivity with an unrelated epitope from the Titin protein presented on cardiac tissue. Here, we investigated the structural mechanism enabling TCR cross-recognition of MAGE-A3 and Titin, and applied the resulting data to rationally design mutants with improved antigen discrimination, providing a proof-of-concept strategy for altering the fine specificity of a TCR towards an intended target antigen. This study represents the first example of direct molecular mimicry leading to clinically relevant fatal toxicity, mediated by a modified enhanced affinity TCR designed for cancer immunotherapy. Furthermore, these data demonstrate that self-antigens that are expressed at high levels on healthy tissue should be treated with extreme caution when designing immuno-therapeutics.


Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cross Reactions/immunology , Immunotherapy/adverse effects , Immunotherapy/methods , Molecular Mimicry , Receptors, Antigen, T-Cell/metabolism , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Cardiotoxicity , Cell Line , Connectin/chemistry , Connectin/immunology , Connectin/metabolism , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Genetic Engineering , Humans , Models, Molecular , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Binding/immunology , Protein Conformation , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
7.
Cancer Immunol Immunother ; 63(5): 437-48, 2014 May.
Article En | MEDLINE | ID: mdl-24531387

Antigen cross-presentation by dendritic cells (DCs) is thought to play a critical role in driving a polyclonal and durable T cell response against cancer. It follows, therefore, that the capacity of emerging immunotherapeutic agents to orchestrate tumour eradication may depend on their ability to induce antigen cross-presentation. ImmTACs [immune-mobilising monoclonal TCRs (T cell receptors) against cancer] are a new class of soluble bi-specific anti-cancer agents that combine pico-molar affinity TCR-based antigen recognition with T cell activation via a CD3-specific antibody fragment. ImmTACs specifically recognise human leucocyte antigen (HLA)-restricted tumour-associated antigens, presented by cancer cells, leading to T cell redirection and a potent anti-tumour response. Using an ImmTAC specific for a HLA-A*02-restricted peptide derived from the melanoma antigen gp100 (termed IMCgp100), we here observe that ImmTAC-driven melanoma-cell death leads to cross-presentation of melanoma antigens by DCs. These, in turn, can activate both melanoma-specific T cells and polyclonal T cells redirected by IMCgp100. Moreover, activation of melanoma-specific T cells by cross-presenting DCs is enhanced in the presence of IMCgp100; a feature that serves to increase the prospect of breaking tolerance in the tumour microenvironment. The mechanism of DC cross-presentation occurs via 'cross-dressing' which involves the rapid and direct capture by DCs of membrane fragments from dying tumour cells. DC cross-presentation of gp100-peptide-HLA complexes was visualised and quantified using a fluorescently labelled soluble TCR. These data demonstrate how ImmTACs engage with the innate and adaptive components of the immune system enhancing the prospect of mediating an effective and durable anti-tumour response in patients.


Cross-Priming/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Receptors, Antigen, T-Cell/therapeutic use , T-Lymphocytes/immunology , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Humans , Melanoma/immunology , Receptors, Antigen, T-Cell/immunology , gp100 Melanoma Antigen/immunology
8.
Sci Transl Med ; 5(197): 197ra103, 2013 Aug 07.
Article En | MEDLINE | ID: mdl-23926201

MAGE A3, which belongs to the family of cancer-testis antigens, is an attractive target for adoptive therapy given its reactivation in various tumors and limited expression in normal tissues. We developed an affinity-enhanced T cell receptor (TCR) directed to a human leukocyte antigen (HLA)-A*01-restricted MAGE A3 antigen (EVDPIGHLY) for use in adoptive therapy. Extensive preclinical investigations revealed no off-target antigen recognition concerns; nonetheless, administration to patients of T cells expressing the affinity-enhanced MAGE A3 TCR resulted in a serious adverse event (SAE) and fatal toxicity against cardiac tissue. We present a description of the preclinical in vitro functional analysis of the MAGE A3 TCR, which failed to reveal any evidence of off-target activity, and a full analysis of the post-SAE in vitro investigations, which reveal cross-recognition of an off-target peptide. Using an amino acid scanning approach, a peptide from the muscle protein Titin (ESDPIVAQY) was identified as an alternative target for the MAGE A3 TCR and the most likely cause of in vivo toxicity. These results demonstrate that affinity-enhanced TCRs have considerable effector functions in vivo and highlight the potential safety concerns for TCR-engineered T cells. Strategies such as peptide scanning and the use of more complex cell cultures are recommended in preclinical studies to mitigate the risk of off-target toxicity in future clinical investigations.


Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Connectin/chemistry , Cross Reactions/immunology , HLA-A1 Antigen/immunology , Neoplasm Proteins/immunology , Peptides/immunology , T-Lymphocytes/metabolism , Amino Acid Sequence , Antigens, Neoplasm/chemistry , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Connectin/immunology , Cross Reactions/drug effects , HEK293 Cells , Humans , Lymphocyte Activation/drug effects , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neoplasm Proteins/chemistry , Peptides/chemistry , Protein Engineering , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects
9.
Oncoimmunology ; 2(11): e26840, 2013 Nov 01.
Article En | MEDLINE | ID: mdl-24482751

Peptide-pulsed T2 cells are routinely used to study T-cell activation by MHC-restricted peptides derived from tumor-associated antigens (TAAs). Nevertheless, the capacity of T2 cells to present antigenic epitopes remains to be precisely quantified, primarily due to the detection limits imposed by available methods. Since naturally-processed TAA-derived epitopes have been shown to be displayed at levels as low as 10-150 copies per cell, highly sensitive detection and quantification techniques are essential to assess the natural degree of T-cell sensitivity. Here, we report the use of soluble, high-affinity T-cell receptors (TCRs) coupled with single-molecule fluorescence microscopy to quantify three reported TAA-derived epitopes on peptide-pulsed T2 cells, dissecting the relationship between concentration of exogenous peptide, number of epitopes presented, and activation of epitope-specific T cells. Our findings indicate that peptide concentrations in the low nanomolar range are required for T2 cells to present TAAs in extents that are comparable to those of malignant cells.

10.
Nat Med ; 18(6): 980-7, 2012 Jun.
Article En | MEDLINE | ID: mdl-22561687

T cell immunity can potentially eradicate malignant cells and lead to clinical remission in a minority of patients with cancer. In the majority of these individuals, however, there is a failure of the specific T cell receptor (TCR)­mediated immune recognition and activation process. Here we describe the engineering and characterization of new reagents termed immune-mobilizing monoclonal TCRs against cancer (ImmTACs). Four such ImmTACs, each comprising a distinct tumor-associated epitope-specific monoclonal TCR with picomolar affinity fused to a humanized cluster of differentiation 3 (CD3)-specific single-chain antibody fragment (scFv), effectively redirected T cells to kill cancer cells expressing extremely low surface epitope densities. Furthermore, these reagents potently suppressed tumor growth in vivo. Thus, ImmTACs overcome immune tolerance to cancer and represent a new approach to tumor immunotherapy.


Cytotoxicity, Immunologic , Neoplasms, Experimental/therapy , Receptors, Antigen, T-Cell/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Immunologic Memory , Immunotherapy , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, SCID , Neoplasms, Experimental/immunology
11.
Traffic ; 12(7): 902-11, 2011 Jul.
Article En | MEDLINE | ID: mdl-21438969

Cytotoxic T lymphocytes (CTL) are potent killers of virally infected and tumorigenic cells. Upon recognition of target cells, CTL undergo polarized secretion of secretory lysosomes at the immunological synapse (IS) that forms between CTL and target. However, the molecular machinery involved in the polarization of secretory lysosomes is still largely uncharacterized. In this paper, we investigated the role of Rab7 in the polarization of secretory lysosomes. We show that silencing of Rab7 by RNA interference reduces the ability of CTL to kill targets. GTP-bound Rab7 and Rab interacting lysosomal protein, RILP, interact and both localize to secretory lysosomes in CTL. Over-expression of RILP recruits dynein to the membranes of secretory lysosomes and triggers their movement toward the centrosome. Together, these results suggest that Rab7 may play a role in secretory lysosome movement toward the centrosome by interacting with RILP to recruit the minus-end motor, dynein.


Secretory Vesicles/metabolism , T-Lymphocytes, Cytotoxic/metabolism , rab GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Dyneins/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins , Mutation , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
12.
J Immunol ; 184(4): 1829-1839, 2010 Feb 15.
Article En | MEDLINE | ID: mdl-20053942

Although several cancer immunotherapy strategies are based on the use of analog peptides and on the modulation of the TCR affinity of adoptively transferred T cells, it remains unclear whether tumor-specific T cell activation by strong and weak TCR stimuli evoke different Ca(2+) signatures from the Ca(2+) intracellular stores and whether the amplitude of Ca(2+) release from the endoplasmic reticulum (ER) can be further modulated by coreceptor binding to peptide/MHC. In this study, we combined functional, structural, and kinetic measurements to correlate the intensity of Ca(2+) signals triggered by the stimulation of the 1G4 T cell clone specific to the tumor epitope NY-ESO-1(157-165). Two analogs of the NY-ESO-1(157-165) peptide, having similar affinity to HLA-A2 molecules, but a 6-fold difference in binding affinity for the 1G4 TCR, resulted in different Ca(2+) signals and T cell activation. 1G4 stimulation by the stronger stimulus emptied the ER of stored Ca(2+), even in the absence of CD8 binding, resulting in sustained Ca(2+) influx. In contrast, the weaker stimulus induced only partial emptying of stored Ca(2+), resulting in significantly diminished and oscillatory Ca(2+) signals, which were enhanced by CD8 binding. Our data define the range of TCR/peptide MHC affinities required to induce depletion of Ca(2+) from intracellular stores and provide insights into the ability of T cells to tailor the use of the CD8 coreceptor to enhance Ca(2+) release from the ER. This, in turn, modulates Ca(2+) influx from the extracellular environment, ultimately controlling T cell activation.


Calcium/metabolism , Endoplasmic Reticulum/metabolism , Epitopes, T-Lymphocyte/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Clone Cells , Crystallography, X-Ray , Cytoplasmic Granules/immunology , Cytoplasmic Granules/metabolism , Cytotoxicity Tests, Immunologic , Endoplasmic Reticulum/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Lymphocyte Activation/immunology , Neoplasm Proteins/metabolism , Peptide Fragments/metabolism , Protein Binding/immunology , Protein Isoforms/immunology , Protein Isoforms/metabolism , Receptors, Antigen, T-Cell/physiology
13.
Traffic ; 9(4): 446-57, 2008 Apr.
Article En | MEDLINE | ID: mdl-18266782

Rab27a is required for polarized secretion of lysosomes from cytotoxic T lymphocytes (CTLs) at the immunological synapse. A series of Rab27a-interacting proteins have been identified; however, only Munc13-4 has been found to be expressed in CTL. In this study, we screened for expression of the synaptotagmin-like proteins (Slps): Slp1/JFC1, Slp2-a/exophilin4, Slp3-a, Slp4/granuphilin, Slp5 and rabphilin in CTL. We found that both Slp1 and Slp2-a are expressed in CTL. Isoforms of Slp2-a in CTL showed variation of the linker region but conserved the C2A and C2B and Slp homology (SHD) domains. Both Slp1 and Slp2-a interact with Rab27a in CTL, and Slp2-a, but not Slp1, is rapidly degraded when Rab27a is absent. Slp2-a contains PEST-like sequences within its linker region, which render it susceptible to degradation. Both Slp1 and Slp2-a localize predominantly to the plasma membrane of both human and mouse CTLs, and we show that Slp2-a can focus tightly at the immunological synapse formed with a target cell. Individual knockouts of either Slp2-a or Slp1 fail to impair CTL-mediated killing of targets; however, overexpression of a dominant-negative construct consisting of the SHD of Slp2-a, which is 56% identical to that of Slp1, reduces target cell death, suggesting that both Slp1 and Slp2-a contribute to secretory lysosome exocytosis from CTL. These results suggest that both Slp1 and Slp2-a may form part of a docking complex, capturing secretory lysosomes at the immunological synapse.


Blood Proteins/immunology , Cell Membrane/metabolism , Membrane Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Blood Proteins/genetics , Calpain/genetics , Calpain/metabolism , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Protein Isoforms/genetics , Protein Isoforms/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vesicular Transport Proteins , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/immunology , rab27 GTP-Binding Proteins
14.
Mol Cancer Ther ; 6(7): 2081-91, 2007 Jul.
Article En | MEDLINE | ID: mdl-17620437

Tumor-associated human telomerase reverse transcriptase (hTERT) is expressed in >85% of human tumors but not in most normal cells. As a result, this antigen has received considerable attention from those interested in cancer immunotherapy. Specifically, there has been strong interest in MHC class I-associated peptides derived from hTERT because these are expressed on the cell surface and thus may enable the targeting of tumor cells. Much of this interest has focused on peptide 540-548, ILAKFLHWL, which was predicted to exhibit the strongest binding to the common HLA A*0201 presenting molecule. The hTERT(540-548) peptide is currently being assessed in therapeutic vaccination trials; however, there is controversy surrounding whether it is naturally processed and presented on the surface of neoplastic cells. Here, we generate two highly sensitive reagents to assess the presentation of hTERT(540-548) on tumor cells: (a) a CD8(+) CTL clone, and (b) a recombinant T-cell receptor (TCR) that binds with picomolar affinity and a half-life exceeding 14 h. This TCR enables the identification of individual HLA A2-hTERT(540-548) complexes on the cell surface. The use of both this TCR and the highly antigen-sensitive CTL clone shows that the hTERT(540-548) peptide cannot be detected on the surface of tumor cells, indicating that this peptide is not a naturally presented epitope. We propose that, in future, rigorous methods must be applied for the validation of peptide epitopes used for clinical applications.


HLA-A Antigens/immunology , Peptide Fragments/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Telomerase/immunology , Amino Acid Sequence , Antigen Presentation/drug effects , Antigen Presentation/immunology , Cell Line, Tumor , Cell Separation , Clone Cells , Enzyme-Linked Immunosorbent Assay , Epitopes , HLA-A2 Antigen , Humans , Interferon-gamma/pharmacology , Molecular Sequence Data , Peptides/chemistry , Peptides/isolation & purification , Proteasome Inhibitors , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/isolation & purification , T-Lymphocytes, Cytotoxic/drug effects , Transfection
15.
J Exp Med ; 204(5): 1131-44, 2007 May 14.
Article En | MEDLINE | ID: mdl-17485514

CD1d-restricted lymphocytes recognize a broad lipid range. However, how CD1d-restricted lymphocytes translate T cell receptor (TCR) recognition of lipids with similar group heads into distinct biological responses remains unclear. Using a soluble invariant NKT (iNKT) TCR and a newly engineered antibody specific for alpha-galactosylceramide (alpha-GalCer)-human CD1d (hCD1d) complexes, we measured the affinity of binding of iNKT TCR to hCD1d molecules loaded with a panel of alpha-GalCer analogues and assessed the rate of dissociation of alpha-GalCer and alpha-GalCer analogues from hCD1d molecules. We extended this analysis by studying iNKT cell synapse formation and iNKT cell activation by the same panel of alpha-GalCer analogues. Our results indicate the unique role of the lipid chain occupying the hCD1d F' channel in modulating TCR binding affinity to hCD1d-lipid complexes, the formation of stable immunological synapse, and cell activation. These data are consistent with previously described conformational changes between empty and loaded hCD1d molecules (Koch, M., V.S. Stronge, D. Shepherd, S.D. Gadola, B. Mathew, G. Ritter, A.R. Fersht, G.S. Besra, R.R. Schmidt, E.Y. Jones, and V. Cerundolo. 2005. Nat. Immunol 6:819-826), suggesting that incomplete occupation of the hCD1d F' channel results in conformational differences at the TCR recognition surface. This indirect effect provides a general mechanism by which lipid-specific lymphocytes are capable of recognizing both the group head and the length of lipid antigens, ensuring greater specificity of antigen recognition.


Antigens, CD1/metabolism , Glycolipids/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Models, Molecular , Receptors, Antigen, T-Cell/metabolism , Antigens, CD1d , Calcium/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Galactosylceramides/immunology , Galactosylceramides/metabolism , Humans , Molecular Structure , Protein Binding
16.
J Cell Sci ; 120(Pt 1): 191-9, 2007 Jan 01.
Article En | MEDLINE | ID: mdl-17164290

Fas ligand (FasL), a potent mediator of apoptosis expressed by CTL and NK cells, is sorted into the inner vesicles of secretory lysosomes for release via exosome-like vesicles. Previous studies identified a proline-rich domain in the cytoplasmic tail required for sorting FasL to secretory lysosomes, but the mechanisms by which this occurs have not been identified. Here we demonstrate that the PRD of FasL binds Fgr, Fyn and Lyn tyrosine kinases, leading to phosphorylation of FasL. Loss of phosphorylation reduces internalisation of FasL into multivesicular bodies. FasL is also directly mono-ubiquitylated at lysines flanking the PRD and mutation of these lysines reduces MVB localisation of FasL. Phosphorylation is not required for ubiquitylation because FasL lacking all tyrosines undergoes mono-ubiquitylation. These studies show that phosphorylation and ubiquitin signals regulate the sorting of FasL to secretory lysosomes by controlling entry into multivesicular bodies.


Fas Ligand Protein/metabolism , Lysosomes/metabolism , Protein Transport/physiology , Secretory Vesicles/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Endosomes/metabolism , Fas Ligand Protein/chemistry , Fas Ligand Protein/genetics , HeLa Cells , Humans , Jurkat Cells , Leukemia, Basophilic, Acute , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn/chemistry , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Rats , Signal Transduction/physiology , src Homology Domains/physiology , src-Family Kinases/chemistry , src-Family Kinases/genetics , src-Family Kinases/metabolism
17.
Nature ; 443(7110): 462-5, 2006 Sep 28.
Article En | MEDLINE | ID: mdl-17006514

Cytotoxic T lymphocytes (CTLs) destroy virally infected and tumorigenic cells by releasing the contents of specialized secretory lysosomes--termed 'lytic granules'--at the immunological synapse formed between the CTL and the target. On contact with the target cell, the microtubule organizing centre of the CTL polarizes towards the target and granules move along microtubules in a minus-end direction towards the polarized microtubule organizing centre. However, the final steps of secretion have remained unclear. Here we show that CTLs do not require actin or plus-end microtubule motors for secretion, but instead the centrosome moves to and contacts the plasma membrane at the central supramolecular activation cluster of the immunological synapse. Actin and IQGAP1 are cleared away from the synapse, and granules are delivered directly to the plasma membrane. These data show that CTLs use a previously unreported mechanism for delivering secretory granules to the immunological synapse, with granule secretion controlled by centrosome delivery to the plasma membrane.


Cell Polarity , Centrosome/metabolism , Cytotoxicity, Immunologic , Secretory Vesicles/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/metabolism , Actins/metabolism , Animals , Biological Transport , Cell Membrane/metabolism , Humans , Mice , Microtubules/metabolism , Molecular Motor Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , ras GTPase-Activating Proteins/metabolism
18.
J Exp Med ; 201(8): 1243-55, 2005 Apr 18.
Article En | MEDLINE | ID: mdl-15837811

Analogue peptides with enhanced binding affinity to major histocompatibility class (MHC) I molecules are currently being used in cancer patients to elicit stronger T cell responses. However, it remains unclear as to how alterations of anchor residues may affect T cell receptor (TCR) recognition. We correlate functional, thermodynamic, and structural parameters of TCR-peptide-MHC binding and demonstrate the effect of anchor residue modifications of the human histocompatibility leukocyte antigens (HLA)-A2 tumor epitope NY-ESO-1(157-165)-SLLMWITQC on TCR recognition. The crystal structure of the wild-type peptide complexed with a specific TCR shows that TCR binding centers on two prominent, sequential, peptide sidechains, methionine-tryptophan. Cysteine-to-valine substitution at peptide position 9, while optimizing peptide binding to the MHC, repositions the peptide main chain and generates subtly enhanced interactions between the analogue peptide and the TCR. Binding analyses confirm tighter binding of the analogue peptide to HLA-A2 and improved soluble TCR binding. Recognition of analogue peptide stimulates faster polarization of lytic granules to the immunological synapse, reduces dependence on CD8 binding, and induces greater numbers of cross-reactive cytotoxic T lymphocyte to SLLMWITQC. These results provide important insights into heightened immunogenicity of analogue peptides and highlight the importance of incorporating structural data into the process of rational optimization of superagonist peptides for clinical trials.


Antigens, Neoplasm/chemistry , Cancer Vaccines/pharmacology , Epitopes, T-Lymphocyte/chemistry , Membrane Proteins/chemistry , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/pharmacology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Cell Line, Tumor , Chemokine CCL4 , Crystallography, X-Ray , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , Immunization , Interferon-gamma/analysis , Macrophage Inflammatory Proteins/analysis , Major Histocompatibility Complex/immunology , Membrane Proteins/immunology , Mice , Mice, Transgenic , Peptides/chemistry , Peptides/immunology , Protein Binding/immunology , Protein Conformation , Receptors, Antigen, T-Cell/chemistry , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Transfection , Vaccines, Synthetic/chemistry
19.
Traffic ; 6(3): 243-51, 2005 Mar.
Article En | MEDLINE | ID: mdl-15702992

Melanocytes and cells of the immune system share an unusual secretory mechanism which uses the lysosome as a regulated secretory organelle. Recently, a number of the proteins required for these 'secretory lysosomes' to undergo exocytosis have been identified. These include Rab27a, Lyst, Rab geranyl geranyl transferase and the adapter protein complex AP-3. Patients lacking any of these proteins are characterized by the rare combination of albinism and immunodeficiency, revealing roles for these proteins in both melanocyte and immune cell secretion. In order to ask how far the link between albinism and immunodeficiency extends we have examined cytotoxic T-lymphocyte (CTL) secretion from two BLOC-3-deficient patients and seven different mouse models of Hermansky-Pudlak syndrome, all of which display defects in pigmentation and platelet function. We find that CTL function is normal in HPS patients and pale-ear mice deficient in BLOC-3, pallid, muted and sandy mice deficient in BLOC-1, ruby-eye mice deficient in BLOC-2 and buff mice deficient in Vps33a. Similarly, the unconventional myosins, Va, VIIa and XV, which can act as effectors for Rab27a in some cell types, are not required in CTL. These results reveal differences in the protein machinery required for biogenesis and/or secretion of lysosome-related organelles in CTL and melanocytes.


Carrier Proteins/physiology , Hermanski-Pudlak Syndrome/physiopathology , Lysosomes/metabolism , Myosins/deficiency , T-Lymphocytes, Cytotoxic/metabolism , Animals , Dyneins/deficiency , Dyneins/physiology , Humans , Melanocytes/metabolism , Mice , Mice, Mutant Strains , Molecular Motor Proteins , Myosin Heavy Chains/deficiency , Myosin Heavy Chains/physiology , Myosin Type V/deficiency , Myosin Type V/physiology , Myosin VIIa , Myosins/physiology , Secretory Vesicles/physiology
20.
Semin Immunol ; 17(1): 87-94, 2005 Feb.
Article En | MEDLINE | ID: mdl-15582491

Cytotoxic T cells (CTL) clear virally infected cells and tumorigenic cells. CTL kill their targets by releasing very potent cytotoxic proteins stored in preformed secretory organelles termed secretory lysosomes. The accurate killing is assured by a vectorial movement of secretory lysosomes along microtubules and focused secretion within the immunological synapse. Some of the proteins controlling these secretion steps have been identified studying CTL from patients affected by severe immunodeficiency. Here we present an over view of how these key regulators such as AP-3 adaptor protein, RabGGTase enzyme, Rab27a small GTP binding protein, Lyst and Munc13-4 act in a very orchestrated way to deliver the "kiss of the death".


GTP-Binding Proteins/immunology , Lysosomes/immunology , Microtubules/immunology , Secretory Vesicles/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Biological Transport, Active/immunology , Humans
...