Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Immunol ; 204(7): 1943-1953, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32102902

ABSTRACT

The molecular rules driving TCR cross-reactivity are poorly understood and, consequently, it is unclear the extent to which TCRs targeting the same Ag recognize the same off-target peptides. We determined TCR-peptide-HLA crystal structures and, using a single-chain peptide-HLA phage library, we generated peptide specificity profiles for three newly identified human TCRs specific for the cancer testis Ag NY-ESO-1157-165-HLA-A2. Two TCRs engaged the same central peptide feature, although were more permissive at peripheral peptide positions and, accordingly, possessed partially overlapping peptide specificity profiles. The third TCR engaged a flipped peptide conformation, leading to the recognition of off-target peptides sharing little similarity with the cognate peptide. These data show that TCRs specific for a cognate peptide recognize discrete peptide repertoires and reconciles how an individual's limited TCR repertoire following negative selection in the thymus is able to recognize a vastly larger antigenic pool.


Subject(s)
HLA-A2 Antigen/immunology , Histocompatibility Antigens/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Cell Line , Humans , Peptide Library
2.
Cent European J Urol ; 71(2): 196-201, 2018.
Article in English | MEDLINE | ID: mdl-30038810

ABSTRACT

INTRODUCTION: Diagnostic pressure on endoscopy suites can result in stent removal not receiving the required priority and unnecessary morbidity for patients. As well as using stents with extraction strings, the introduction of a portable single-use flexible cystoscope for ureteric stent removal (Isiris™), offered an opportunity to negotiate these issues by relocating stent removal to the office/clinic. This study aimed to determine whether such flexibility reduced stent dwell time with the assumption this would improve patient experience and decrease associated complications. MATERIALS AND METHODS: A retrospective review of ureteric stents placed during stone procedures was undertaken. Data collection included; patient demographics; stent dwell times; the number of emergency department (ED) attendances and hospital readmissions; procedure cancellation rates and the number of urinary tract infections. RESULTS: In total, 162 stents were removed (113 Standard, 34 Isiris™, 15 via strings). Excess dwell time was reduced in both Isiris™ (median 1 day, mean 1.37 days, p = 0.0009) and Strings Groups (median 0.96 days, mean 0.96 days, p = 0.022) compared with the Standard Group (median 8 days, mean 15.34 days).ED attendances and readmissions were reduced by 33.5% and 22% respectively in the Isiris™ Group compared with the Standard Group. There were no ED attendances in the Strings Group. Reductions in length of stay, urine infections and cancellation on the day of procedures were also observed. CONCLUSIONS: The clinical flexibility provided by Isiris™ and 'stents on strings' has objectively improved patient experience and is associated with a reduction in complications as well as increasing diagnostic capacity and cost efficacy.

3.
Sci Rep ; 6: 18851, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26758806

ABSTRACT

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.


Subject(s)
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
4.
BMJ Case Rep ; 20122012 Nov 11.
Article in English | MEDLINE | ID: mdl-23148399

ABSTRACT

We describe a case of methaemoglobinaemia (MtHb) in a previously healthy 39-year-old gentleman who presented with a traumatic glass laceration to his right wrist that required emergency surgery to control bleeding and repair his ulnar artery. The MtHb was noted on blood gas analysis by the anaesthetist after the patient had a drop in arterial oxygen saturation under general anaesthetic. We initially suspected the lidocaine local anaesthetic injected proximal to his wound for pain control in the emergency department an hour preoperatively, but then discovered that the patient was a recreational user of 'poppers' and had in fact been using these drugs just before his injury and hospitalisation. The patient's condition stabilised overnight with conservative management. Given how commonly hand surgeons and other clinical staff use local anaesthetics, we reviewed the literature on this uncommon, but potentially fatal, complication, its causes and evidence-based management.


Subject(s)
Inhalant Abuse/complications , Inhalant Abuse/diagnosis , Intraoperative Complications/chemically induced , Intraoperative Complications/diagnosis , Methemoglobinemia/chemically induced , Methemoglobinemia/diagnosis , Nitrites/toxicity , Wounds, Penetrating/surgery , Wrist Injuries/surgery , Adult , Diagnosis, Differential , Humans , Male
5.
Nat Med ; 18(6): 980-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22561687

ABSTRACT

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.


Subject(s)
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
6.
Protein Cell ; 1(12): 1118-27, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21213105

ABSTRACT

Using directed mutagenesis and phage display on a soluble fragment of the human immunoglobulin super-family receptor ILT2 (synonyms: LIR1, MIR7, CD85j), we have selected a range of mutants with binding affinities enhanced by up to 168,000-fold towards the conserved region of major histocompatibility complex (MHC) class I molecules. Produced in a dimeric form, either by chemical cross-linking with bivalent polyethylene glycol (PEG) derivatives or as a genetic fusion with human IgG Fc-fragment, the mutants exhibited a further increase in ligand-binding strength due to the avidity effect, with resident half-times (t(1/2)) on the surface of MHC I-positive cells of many hours. The novel compounds antagonized the interaction of CD8 co-receptor with MHC I in vitro without affecting the peptide-specific binding of T-cell receptors (TCRs). In both cytokine-release assays and cell-killing experiments the engineered receptors inhibited the activation of CD8(+) cytotoxic T lymphocytes (CTLs) in the presence of their target cells, with subnanomolar potency and in a dose-dependent manner. As a selective inhibitor of CD8(+) CTL responses, the engineered high affinity ILT2 receptor presents a new tool for studying the activation mechanism of different subsets of CTLs and could have potential for the development of novel autoimmunity therapies.


Subject(s)
Antigens, CD/genetics , Antigens, CD/pharmacology , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Lymphocyte Activation/immunology , Receptors, Immunologic/genetics , Amino Acid Sequence , Antigens, CD/chemistry , Autoimmunity , Biological Assay , Cell Line , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Dose-Response Relationship, Immunologic , Humans , Immunoglobulins/immunology , Immunoglobulins/metabolism , Immunologic Factors/chemistry , Kinetics , Leukocyte Immunoglobulin-like Receptor B1 , Lymphocyte Activation/genetics , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Molecular Sequence Data , Molecular Targeted Therapy , Mutagenesis, Site-Directed , Peptide Library , Polyethylene Glycols , Protein Binding/genetics , Protein Binding/immunology , Receptors, Immunologic/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
7.
Mol Cancer Ther ; 6(7): 2081-91, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620437

ABSTRACT

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.


Subject(s)
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
8.
Protein Eng Des Sel ; 20(8): 397-403, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17644531

ABSTRACT

Naturally selected T-cell receptors (TCRs) are characterised by low binding affinities, typically in the range 1-100 microM. Crystal structures of syngeneic TCRs bound to peptide major histocompatibility complex (pMHC) antigens exhibit a conserved mode of binding characterised by a distinct diagonal binding geometry, with poor shape complementarity (SC) between receptor and ligand. Here, we report the structures of three in vitro affinity enhanced TCRs that recognise the pMHC tumour epitope NY-ESO(157-165) (SLLMWITQC). These crystal structures reveal that the docking mode for the high affinity TCRs is identical to that reported for the parental wild-type TCR, with only subtle changes in the mutated complementarity determining regions (CDRs) that form contacts with pMHC; both CDR2 and CDR3 mutations act synergistically to improve the overall affinity. Comparison of free and bound TCR structures for both wild-type and a CDR3 mutant reveal an induced fit mechanism arising from restructuring of CDR3 loops which allows better peptide binding. Overall, an increased interface area, improved SC and additional H-bonding interactions are observed, accounting for the increase in affinity. Most notably, there is a marked increase in the SC for the central methionine and tryptophan peptide motif over the native TCR.


Subject(s)
Crystallography, X-Ray , Major Histocompatibility Complex/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Escherichia coli/genetics , Humans , Hydrogen Bonding , Kinetics , Ligands , Models, Molecular , Mutation , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , Surface Plasmon Resonance
9.
Protein Sci ; 15(4): 710-21, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16600963

ABSTRACT

The mammalian alpha/beta T cell receptor (TCR) repertoire plays a pivotal role in adaptive immunity by recognizing short, processed, peptide antigens bound in the context of a highly diverse family of cell-surface major histocompatibility complexes (pMHCs). Despite the extensive TCR-MHC interaction surface, peptide-independent cross-reactivity of native TCRs is generally avoided through cell-mediated selection of molecules with low inherent affinity for MHC. Here we show that, contrary to expectations, the germ line-encoded complementarity determining regions (CDRs) of human TCRs, namely the CDR2s, which appear to contact only the MHC surface and not the bound peptide, can be engineered to yield soluble low nanomolar affinity ligands that retain a surprisingly high degree of specificity for the cognate pMHC target. Structural investigation of one such CDR2 mutant implicates shape complementarity of the mutant CDR2 contact interfaces as being a key determinant of the increased affinity. Our results suggest that manipulation of germ line CDR2 loops may provide a useful route to the production of high-affinity TCRs with therapeutic and diagnostic potential.


Subject(s)
Complementarity Determining Regions/chemistry , Peptides/metabolism , Receptors, Antigen, T-Cell/chemistry , Antigens/metabolism , Cell Line, Transformed , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Crystallography, X-Ray , Humans , Kinetics , Ligands , Major Histocompatibility Complex , Models, Molecular , Mutation , Nerve Tissue Proteins , Peptide Library , Peptides/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta , Sensitivity and Specificity , Substrate Specificity , Surface Plasmon Resonance
10.
Nat Biotechnol ; 23(3): 349-54, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15723046

ABSTRACT

Peptides derived from almost all proteins, including disease-associated proteins, can be presented on the cell surface as peptide-human leukocyte antigen (pHLA) complexes. T cells specifically recognize pHLA with their clonally rearranged T-cell receptors (TCRs), whose natural affinities are limited to approximately 1-100 muM. Here we describe the display of ten different human TCRs on the surface of bacteriophage, stabilized by a nonnative interchain disulfide bond. We report the directed evolution of high-affinity TCRs specific for two different pHLAs: the human T-cell lymphotropic virus type 1 (HTLV-1) tax(11-19) peptide-HLA-A(*)0201 complex and the NY-ESO-1(157-165) tumor-associated peptide antigen-HLA-A(*)0201 complex, with affinities of up to 2.5 nM and 26 pM, respectively, and we demonstrate their high specificity and sensitivity for targeting of cell-surface pHLAs.


Subject(s)
Antibody Affinity , Antibody Formation , Complementarity Determining Regions/genetics , Directed Molecular Evolution/methods , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/immunology , Microchemistry/methods , Peptide Library , Protein Engineering/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Humans , Immunoglobulin Fab Fragments/genetics , Protein Binding , Receptors, Antigen, T-Cell/biosynthesis , Recombination, Genetic/genetics
11.
Protein Eng ; 16(9): 707-11, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14560057

ABSTRACT

Antibody and T-cell receptors (TCRs) are the primary recognition molecules of the adaptive immune system. Antibodies have been extensively characterized and are being developed for a large number of therapeutic applications. This has been possible because of the ability to manufacture stable, soluble, monoclonal antibodies which retain the antigen specificity of B cells. Unlike antibodies, TCRs are not expressed in a soluble form, but are anchored to the T-cell surface by an insoluble trans-membrane domain. Characterization and development of TCRs has been hampered by the lack of suitable methods for producing them as soluble and stable proteins. Here we report the engineering of soluble human TCRs suitable for crystallization studies and potentially for in vivo therapeutic use.


Subject(s)
Receptors, Antigen, T-Cell/chemistry , T-Lymphocytes/chemistry , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Protein Structure, Tertiary/physiology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...