Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
PLoS Pathog ; 15(2): e1007567, 2019 02.
Article in English | MEDLINE | ID: mdl-30789961

ABSTRACT

Most studies of T lymphocytes focus on recognition of classical major histocompatibility complex (MHC) class I or II molecules presenting oligopeptides, yet there are numerous variations and exceptions of biological significance based on recognition of a wide variety of nonclassical MHC molecules. These include αß and γδ T cells that recognize different class Ib molecules (CD1, MR-1, HLA-E, G, F, et al.) that are nearly monomorphic within a given species. Collectively, these T cells can be considered "unconventional," in part because they recognize lipids, metabolites, and modified peptides. Unlike classical MHC-specific cells, unconventional T cells generally exhibit limited T-cell antigen receptor (TCR) repertoires and often produce innate immune cell-like rapid effector responses. Exploiting this system in new generation vaccines for human immunodeficiency virus (HIV), tuberculosis (TB), other infectious agents, and cancer was the focus of a recent workshop, "Immune Surveillance by Non-classical MHC Molecules: Improving Diversity for Antigens," sponsored by the National Institute of Allergy and Infectious Diseases. Here, we summarize salient points presented regarding the basic immunobiology of unconventional T cells, recent advances in methodologies to measure unconventional T-cell activity in diseases, and approaches to harness their considerable clinical potential.


Subject(s)
Immunologic Surveillance/immunology , Major Histocompatibility Complex/immunology , Animals , Antigens , HLA Antigens , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Receptors, Antigen, T-Cell , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology
3.
PLoS One ; 13(10): e0205491, 2018.
Article in English | MEDLINE | ID: mdl-30321203

ABSTRACT

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.


Subject(s)
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 in English | MEDLINE | ID: mdl-28640942

ABSTRACT

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.


Subject(s)
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 in English | MEDLINE | ID: mdl-27401039

ABSTRACT

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.


Subject(s)
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 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
7.
Mol Ther ; 23(9): 1541-50, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25896248

ABSTRACT

Here, we describe a fatal serious adverse event observed in a patient infused with autologous T-cell receptor (TCR) transduced T cells. This TCR, originally obtained from a melanoma patient, recognizes the well-described HLA-A*0201 restricted 26-35 epitope of MART-1, and was not affinity enhanced. Patient 1 with metastatic melanoma experienced a cerebral hemorrhage, epileptic seizures, and a witnessed cardiac arrest 6 days after cell infusion. Three days later, the patient died from multiple organ failure and irreversible neurologic damage. After T-cell infusion, levels of IL-6, IFN-γ, C-reactive protein (CRP), and procalcitonin increased to extreme levels, indicative of a cytokine release syndrome or T-cell-mediated inflammatory response. Infused T cells could be recovered from blood, broncho-alveolar lavage, ascites, and after autopsy from tumor sites and heart tissue. High levels of NT-proBNP indicate semi-acute heart failure. No cross reactivity of the modified T cells toward a beating cardiomyocyte culture was observed. Together, these observations suggest that high levels of inflammatory cytokines alone or in combination with semi-acute heart failure and epileptic seizure may have contributed substantially to the occurrence of the acute and lethal event. Protocol modifications to limit the risk of T-cell activation-induced toxicity are discussed.


Subject(s)
Cell- and Tissue-Based Therapy/adverse effects , Immunotherapy, Adoptive/adverse effects , MART-1 Antigen/immunology , Receptors, Antigen, T-Cell/genetics , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adult , Cell- and Tissue-Based Therapy/methods , Fatal Outcome , Female , Humans , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , MART-1 Antigen/metabolism , Melanoma/diagnosis , Melanoma/genetics , Melanoma/immunology , Melanoma/therapy , Neoplasm Staging , Receptors, Antigen, T-Cell/metabolism , Transduction, Genetic
8.
Mol Immunol ; 67(2 Pt A): 67-74, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25708206

ABSTRACT

Overcoming immunosuppression and activating a cytotoxic T cell response has the potential to halt the progression of cancer and, in some circumstances, eradicate it. Designing therapeutic interventions that achieve this goal has proven challenging, but now a greater understanding of the complexities of immune responses is beginning to produce some notable breakthroughs. ImmTACs (immune-mobilising monoclonal TCRs against cancer) are a new class of bispecific reagents, based on soluble monoclonal T cell receptors, which have been engineered to possess extremely high affinity for cognate tumour antigen. In this way, ImmTACs overcome the problem of low affinity tumour-specific T cells imposed by thymic selection and provide access to the large number of antigens presented as peptide-HLA complexes. Once bound to tumour cells the anti-CD3 effector end of the ImmTAC drives recruitment of polyclonal T cells to the tumour site, leading to a potent redirected T cell response and tumour cell destruction. Extensive in vitro testing coupled with promising early clinical data has provided an enhanced appreciation of ImmTAC function in vivo and indicates their potential therapeutic benefit in terms of a durable response and ultimately the breaking of T cell tolerance. This review introduces ImmTACs in the context of immunotherapy, and outlines their design, construction and mechanism of action, as well as examining target selection and aspects of preclinical safety testing.


Subject(s)
Antibodies, Bispecific/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Antibodies, Bispecific/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Humans , Models, Immunological , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Reproducibility of Results , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
9.
Cancer Immunol Immunother ; 63(5): 437-48, 2014 May.
Article in English | MEDLINE | ID: mdl-24531387

ABSTRACT

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.


Subject(s)
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
10.
Sci Transl Med ; 5(197): 197ra103, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23926201

ABSTRACT

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.


Subject(s)
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
11.
Blood ; 122(6): 863-71, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23770775

ABSTRACT

An obstacle to cancer immunotherapy has been that the affinity of T-cell receptors (TCRs) for antigens expressed in tumors is generally low. We initiated clinical testing of engineered T cells expressing an affinity-enhanced TCR against HLA-A*01-restricted MAGE-A3. Open-label protocols to test the TCRs for patients with myeloma and melanoma were initiated. The first two treated patients developed cardiogenic shock and died within a few days of T-cell infusion, events not predicted by preclinical studies of the high-affinity TCRs. Gross findings at autopsy revealed severe myocardial damage, and histopathological analysis revealed T-cell infiltration. No MAGE-A3 expression was detected in heart autopsy tissues. Robust proliferation of the engineered T cells in vivo was documented in both patients. A beating cardiomyocyte culture generated from induced pluripotent stem cells triggered T-cell killing, which was due to recognition of an unrelated peptide derived from the striated muscle-specific protein titin. These patients demonstrate that TCR-engineered T cells can have serious and not readily predictable off-target and organ-specific toxicities and highlight the need for improved methods to define the specificity of engineered TCRs.


Subject(s)
Cardiovascular Diseases/complications , Melanoma/blood , Multiple Myeloma/blood , Muscle Proteins/metabolism , Myocardium/pathology , Protein Kinases/metabolism , T-Lymphocytes/cytology , Alleles , Amino Acid Motifs , Antigens, Neoplasm/metabolism , Cell Culture Techniques , Connectin , Cytokines/metabolism , Epitopes/metabolism , HLA-A Antigens/metabolism , Humans , Immunotherapy, Adoptive , Induced Pluripotent Stem Cells/cytology , Male , Melanoma/therapy , Middle Aged , Multiple Myeloma/therapy , Myocardium/immunology , Neoplasm Proteins/metabolism , Peptides/metabolism , Protein Engineering , Receptors, Antigen, T-Cell/immunology
12.
Cancer Immunol Immunother ; 62(4): 773-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23263452

ABSTRACT

NY-ESO-1 and LAGE-1 are cancer testis antigens with an ideal profile for tumor immunotherapy, combining up-regulation in many cancer types with highly restricted expression in normal tissues and sharing a common HLA-A*0201 epitope, 157-165. Here, we present data to describe the specificity and anti-tumor activity of a bifunctional ImmTAC, comprising a soluble, high-affinity T-cell receptor (TCR) specific for NY-ESO-1157-165 fused to an anti-CD3 scFv. This reagent, ImmTAC-NYE, is shown to kill HLA-A2, antigen-positive tumor cell lines, and freshly isolated HLA-A2- and LAGE-1-positive NSCLC cells. Employing time-domain optical imaging, we demonstrate in vivo targeting of fluorescently labelled high-affinity NYESO-specific TCRs to HLA-A2-, NY-ESO-1157-165-positive tumors in xenografted mice. In vivo ImmTAC-NYE efficacy was tested in a tumor model in which human lymphocytes were stably co-engrafted into NSG mice harboring tumor xenografts; efficacy was observed in both tumor prevention and established tumor models using a GFP fluorescence readout. Quantitative RT-PCR was used to analyze the expression of both NY-ESO-1 and LAGE-1 antigens in 15 normal tissues, 5 cancer cell lines, 10 NSCLC, and 10 ovarian cancer samples. Overall, LAGE-1 RNA was expressed at a greater frequency and at higher levels than NY-ESO-1 in the tumor samples. These data support the clinical utility of ImmTAC-NYE as an immunotherapeutic agent for a variety of cancers.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Membrane Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antigens, Neoplasm/biosynthesis , Antigens, Surface/biosynthesis , CD3 Complex/immunology , Cell Line, Tumor , Epitopes/immunology , Female , HLA-A2 Antigen/immunology , Humans , Immunoglobulin Fragments/immunology , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Melanoma/immunology , Melanoma/metabolism , Membrane Proteins/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Random Allocation , Recombinant Fusion Proteins/immunology , Xenograft Model Antitumor Assays
13.
Oncoimmunology ; 2(11): e26840, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24482751

ABSTRACT

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.

14.
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
15.
Protein Expr Purif ; 76(2): 165-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20709174

ABSTRACT

DNA topoisomerase type II enzymes are well-validated targets of anti-bacterial and anti-cancer compounds. In order to facilitate discovery of these types of inhibitors human topoisomerase II in vitro assays can play an important role to support drug discovery processes. Typically, human topoisomerase IIα proteins have been purified from human cell lines or as untagged proteins from yeast cells. This study reports a method for the rapid over-expression and purification of active GST-tagged human topoisomerase IIα using the baculovirus mediated insect cell expression system. Expression of the GST fused protein was observed in the nuclear fraction of insect cells. High yields (40 mg/L i.e. 8 mg/10(9) cells) at >80% purity of this target was achieved by purification using a GST HiTrap column followed by size exclusion chromatography. Functional activity of GST-tagged human topoisomerase IIα was demonstrated by ATP-dependent relaxation of supercoiled DNA in an agarose gel based assay. An 8-fold DNA-dependent increase in ATPase activity of this target compared to its intrinsic activity was also demonstrated in a high-throughput ATPase fluorescence based assay. Human topoisomerase IIα inhibitors etoposide, quercetin and suramin were tested in the fluorescence assay. IC(50) values obtained were in good agreement with published data. These inhibitors also demonstrated ≥ 30-fold potency over the anti-bacterial topoisomerase II inhibitor ciprofloxacin in the assay. Collectively these data validated the enzyme and the high-throughput fluorescence assay as tools for inhibitor identification and selectivity studies.


Subject(s)
Antigens, Neoplasm/isolation & purification , Cloning, Molecular/methods , DNA Topoisomerases, Type II/isolation & purification , DNA-Binding Proteins/isolation & purification , High-Throughput Screening Assays/methods , Recombinant Fusion Proteins/isolation & purification , Adenosine Diphosphate/metabolism , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Baculoviridae/genetics , DNA Topoisomerases, Type II/biosynthesis , DNA Topoisomerases, Type II/genetics , DNA, Circular/chemistry , DNA, Circular/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Enzyme Inhibitors , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Inhibitory Concentration 50 , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Spectrometry, Fluorescence , Spodoptera/metabolism
16.
Expert Opin Drug Discov ; 6(12): 1215-25, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22647062

ABSTRACT

INTRODUCTION: High-throughput screening (HTS) has been and is likely to remain one of the most widely used tools for Hit identification in the pharmaceutical, biotechnology and academic sectors. It has evolved into a highly integrated and automated process enabling the screening of millions of compounds in a timely manner. It is of paramount importance that appropriate biological reagents are utilized in an HTS campaign as their quality and physiological relevance will influence the likelihood of the activities of any identified Hits translating in vivo. AREAS COVERED: This article covers the strategies that can be used to efficiently design and generate biological reagents for the development of kinase assays and their subsequent use in HTS campaigns. The authors describe the variety of molecular biology and expression methodologies available to yield biological reagents of high quality, physiological relevance and amenable to kinase drug discovery. EXPERT OPINION: The techniques now available for gene cloning and protein expression are vast and can be overwhelming. Therefore, we provide guidelines for the most effective route to generate high quality, physiologically relevant biological reagents for kinase drug discovery. The methods available for the generation of biological reagents have undergone significant advances and some of these have been driven by the requirements of HTS campaigns. If the approaches described herein are implemented, it is anticipated they will result in the generation of suitable biological reagents for the development of kinase assays for HTS campaigns.

17.
Biochem J ; 419(1): 65-73, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19061480

ABSTRACT

Identification of small-molecule inhibitors by high-throughput screening necessitates the development of robust, reproducible and cost-effective assays. The assay approach adopted may utilize isolated proteins or whole cells containing the target of interest. To enable protein-based assays, the baculovirus expression system is commonly used for generation and isolation of recombinant proteins. We have applied the baculovirus system into a cell-based assay format using NIK [NF-kappaB (nuclear factor kappaB)-inducing kinase] as a paradigm. We illustrate the use of the insect-cell-based assay in monitoring the activity of NIK against its physiological downstream substrate IkappaB (inhibitor of NF-kappaB) kinase-1. The assay was robust, yielding a signal/background ratio of 2:1 and an average Z' value of >0.65 when used to screen a focused compound set. Using secondary assays to validate a selection of the hits, we identified a compound that (i) was non-cytotoxic, (ii) interacted directly with NIK, and (iii) inhibited lymphotoxin-induced NF-kappaB p52 translocation to the nucleus. The insect cell assay represents a novel approach to monitoring kinase inhibition, with major advantages over other cell-based systems including ease of use, amenability to scale-up, protein expression levels and the flexibility to express a number of proteins by infecting with numerous baculoviruses.


Subject(s)
Biological Assay/methods , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Cell Line , Humans , I-kappa B Kinase/metabolism , Models, Biological , NF-kappa B/metabolism , NF-kappa B p52 Subunit/metabolism , Phosphorylation , Spodoptera , NF-kappaB-Inducing Kinase
19.
Mol Cell Biol ; 26(17): 6727-38, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16914752

ABSTRACT

Deciphering the role of lymphocyte membrane proteins depends on dissecting the role of a protein in the steady state and on engagement with its ligand. We show that expression of CD6 in T cells limits their responsiveness but that engagement by the physiological ligand CD166 gives costimulation. This costimulatory effect of CD6 is mediated through phosphorylation-dependent binding of a specific tyrosine residue, 662Y, in its cytoplasmic region to the adaptor SLP-76. A direct interaction between SLP-76 and CD6 was shown by binding both to a phosphorylated peptide (equilibrium dissociation constant [K(D)] = 0.5 muM at 37 degrees C) and, using a novel approach, to native phosphorylated CD6. Evidence that CD6 and SLP-76 interact in cells was obtained in coprecipitation experiments with normal human T cells. Analysis of human CD6 mutants in a murine T-cell hybridoma model showed that both costimulation by CD6 and the interaction between CD6 and SLP-76 were dependent on 662Y. The results have implications for regulation by CD6 and the related T-cell surface protein, CD5.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , Phosphoproteins/metabolism , T-Lymphocytes/immunology , Activated-Leukocyte Cell Adhesion Molecule/immunology , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/chemistry , Antigens, Differentiation, T-Lymphocyte/chemistry , CD5 Antigens/chemistry , Humans , Interleukin-2/biosynthesis , Jurkat Cells , Mice , Peptides/metabolism , Phosphorylation , Protein Binding , src Homology Domains
20.
Protein Expr Purif ; 47(2): 591-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16481201

ABSTRACT

Human BCRP and OATP1B1 have recently been identified as important transporters in the absorption, distribution, and elimination of clinically significant drugs. In this report, we illustrate the use of modified baculoviruses, termed BacMam viruses for the expression of functional BCRP and OATP1B1 in mammalian cells. We show a variety of host cells efficiently transduced to express BCRP including HEK 293, LLC-PK, and U-2 OS, where protein levels on the cell-surface were modulated by titrating different amounts of viral inoculum. In addition, using the BODIPY-prazosin efflux assay and the BacMam reagent we illustrate inhibition of BCRP activity with GF120918 or Fumitremorgin C. Furthermore, we present data demonstrating simultaneous expression of BCRP and OATP1B1 in BacMam transduced mammalian cells by simply adding viral inoculum of each transporter. Thus these results indicate that BacMam mediated gene delivery provides a novel and efficient research tool for the investigation of single or multiple transporters in vitro.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Baculoviridae , Cell Membrane/metabolism , Neoplasm Proteins/biosynthesis , Organic Anion Transporters/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Acridines/pharmacology , Animals , COS Cells , Cell Line, Tumor , Cell Membrane/genetics , Chlorocebus aethiops , Humans , Indoles/pharmacology , Liver-Specific Organic Anion Transporter 1 , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Organic Anion Transporters/genetics , Spodoptera/cytology , Swine , Tetrahydroisoquinolines/pharmacology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...