Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 447
Filter
1.
Nature ; 628(8007): 416-423, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538786

ABSTRACT

Antibody and chimeric antigen receptor (CAR) T cell-mediated targeted therapies have improved survival in patients with solid and haematologic malignancies1-9. Adults with T cell leukaemias and lymphomas, collectively called T cell cancers, have short survival10,11 and lack such targeted therapies. Thus, T cell cancers particularly warrant the development of CAR T cells and antibodies to improve patient outcomes. Preclinical studies showed that targeting T cell receptor ß-chain constant region 1 (TRBC1) can kill cancerous T cells while preserving sufficient healthy T cells to maintain immunity12, making TRBC1 an attractive target to treat T cell cancers. However, the first-in-human clinical trial of anti-TRBC1 CAR T cells reported a low response rate and unexplained loss of anti-TRBC1 CAR T cells13,14. Here we demonstrate that CAR T cells are lost due to killing by the patient's normal T cells, reducing their efficacy. To circumvent this issue, we developed an antibody-drug conjugate that could kill TRBC1+ cancer cells in vitro and cure human T cell cancers in mouse models. The anti-TRBC1 antibody-drug conjugate may provide an optimal format for TRBC1 targeting and produce superior responses in patients with T cell cancers.


Subject(s)
Immunoconjugates , Leukemia, T-Cell , Lymphoma, T-Cell , Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocytes , Animals , Female , Humans , Mice , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Immunotherapy, Adoptive , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/immunology , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
2.
Sci Rep ; 12(1): 303, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34996924

ABSTRACT

Targeting the programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) axis with monoclonal antibodies (mAbs) represents a crucial breakthrough in anticancer therapy, but mAbs are limited by their poor oral bioavailability, adverse events in multiple organ systems, and primary, adaptive, and acquired resistance, amongst other issues. More recently, the advent of small molecule inhibitors that target the PD-1/PD-L1 axis have shown promising cellular inhibitory activity and the potential to counteract the disadvantages of mAbs. In this study, structure-based virtual screening identified small molecule inhibitors that effectively inhibited the PD-1/PD-L1 interaction. Six of those small molecule inhibitors were applied to cell-based experiments targeting PD-1: CH-1, CH-2, CH-3, CH-4, CH-5, and CH-6. Of all 6, CH-4 displayed the lowest cytotoxicity and strongest inhibitory activity towards the PD-1/PD-L1 interaction. The experiments revealed that CH-4 inhibited the interaction of soluble form PD-L1 (sPD-L1) with PD-1 surface protein expressed by KG-1 cells. Investigations into CH-4 analogs revealed that CH-4.7 effectively blocked the PD-1/sPD-L1 interaction, but sustained the secretion of interleukin-2 and interferon-γ by Jurkat cells. Our experiments revealed a novel small molecule inhibitor that blocks the interaction of PD-1/sPD-L1 and potentially offers an alternative PD-1 target for immune checkpoint therapy.


Subject(s)
B7-H1 Antigen/metabolism , Immune Checkpoint Inhibitors/pharmacology , Leukemia, T-Cell/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Computer-Aided Design , Drug Design , HEK293 Cells , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/immunology , Leukemia, T-Cell/metabolism , Molecular Docking Simulation , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/metabolism , Protein Conformation , Structure-Activity Relationship
3.
Blood Cancer J ; 11(6): 119, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162832

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy is the most active field in immuno-oncology and brings substantial benefit to patients with B cell malignancies. However, the complex procedure for CAR T-cell generation hampers its widespread applications. Here, we describe a novel approach in which human CAR T cells can be generated within the host upon injecting an Adeno-associated virus (AAV) vector carrying the CAR gene, which we call AAV delivering CAR gene therapy (ACG). Upon single infusion into a humanized NOD.Cg-Prkdcscid Il2rgem26/Nju tumor mouse model of human T-cell leukemia, AAV generates sufficient numbers of potent in vivo CAR cells, resulting in tumor regression; these in vivo-generated CAR cells produce antitumor immunological characteristics. This instantaneous generation of in vivo CAR T cells may bypass the need for patient lymphodepletion, as well as the ß processes of traditional CAR T-cell production, which may make CAR therapy simpler and less expensive. It may allow the development of intricate, individualized treatments in the form of on-demand and diverse therapies.


Subject(s)
Dependovirus , Genetic Therapy , Genetic Vectors , Leukemia, T-Cell , Receptors, Chimeric Antigen , Transduction, Genetic , Animals , HEK293 Cells , Humans , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/immunology , Leukemia, T-Cell/therapy , Mice , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Xenograft Model Antitumor Assays
4.
Leukemia ; 35(12): 3466-3481, 2021 12.
Article in English | MEDLINE | ID: mdl-34035409

ABSTRACT

Targeting T cell malignancies using chimeric antigen receptor (CAR) T cells is hindered by 'T v T' fratricide against shared antigens such as CD3 and CD7. Base editing offers the possibility of seamless disruption of gene expression of problematic antigens through creation of stop codons or elimination of splice sites. We describe the generation of fratricide-resistant T cells by orderly removal of TCR/CD3 and CD7 ahead of lentiviral-mediated expression of CARs specific for CD3 or CD7. Molecular interrogation of base-edited cells confirmed elimination of chromosomal translocations detected in conventional Cas9 treated cells. Interestingly, 3CAR/7CAR co-culture resulted in 'self-enrichment' yielding populations 99.6% TCR-/CD3-/CD7-. 3CAR or 7CAR cells were able to exert specific cytotoxicity against leukaemia lines with defined CD3 and/or CD7 expression as well as primary T-ALL cells. Co-cultured 3CAR/7CAR cells exhibited highest cytotoxicity against CD3 + CD7 + T-ALL targets in vitro and an in vivo human:murine chimeric model. While APOBEC editors can reportedly exhibit guide-independent deamination of both DNA and RNA, we found no problematic 'off-target' activity or promiscuous base conversion affecting CAR antigen-specific binding regions, which may otherwise redirect T cell specificity. Combinational infusion of fratricide-resistant anti-T CAR T cells may enable enhanced molecular remission ahead of allo-HSCT for T cell malignancies.


Subject(s)
Antigens, CD7/genetics , CD3 Complex/genetics , Immunotherapy, Adoptive/methods , Leukemia, T-Cell/therapy , T-Lymphocytes/immunology , Animals , Antigens, CD7/chemistry , Antigens, CD7/metabolism , CD3 Complex/antagonists & inhibitors , CD3 Complex/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Female , Gene Editing , Humans , Leukemia, T-Cell/immunology , Leukemia, T-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
5.
Mol Cancer ; 19(1): 162, 2020 11 21.
Article in English | MEDLINE | ID: mdl-33218364

ABSTRACT

Targeting T cell receptor ß-chain constant region 1 (TRBC1) CAR-T could specifically kill TRBC1+ T-cell malignancies. However, over-expressed CARs on anti-TRBC1 CAR transduced TRBC1+ T cells (CAR-C1) bound to autologous TRBC1, masking TRBC1 from identification by other anti-TRBC1 CAR-T, and moreover only the remaining unoccupied CARs recognized TRBC1+ cells, considerably reducing therapeutic potency of CAR-C1. In addition, co-culture of anti-TRBC1 CAR-T and TRBC1+ cells could promote exhaustion and terminal differentiation of CAR-T. These findings provide a rationale for pre-depleting TRBC1+ T cells before anti-TRBC1 CAR-T manufacturing.


Subject(s)
Cytotoxicity, Immunologic/immunology , Immunotherapy, Adoptive/methods , Leukemia, T-Cell/therapy , Lymphocyte Depletion/methods , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Apoptosis , Cell Proliferation , Humans , Leukemia, T-Cell/immunology , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Chimeric Antigen/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cancer Res ; 80(11): 2298-2310, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32156778

ABSTRACT

Immunotherapies targeting programmed cell death protein 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) immune checkpoints represent a major breakthrough in cancer treatment. PD-1 is an inhibitory receptor expressed on the surface of activated T cells that dampens T-cell receptor (TCR)/CD28 signaling by engaging with its ligand PD-L1 expressed on cancer cells. Despite the clinical success of PD-1 blockade using mAbs, most patients do not respond to the treatment, and the underlying regulatory mechanisms of PD-1 remain incompletely defined. Here we show that PD-1 is extensively N-glycosylated in T cells and the intensities of its specific glycoforms are altered upon TCR activation. Glycosylation was critical for maintaining PD-1 protein stability and cell surface localization. Glycosylation of PD-1, especially at the N58 site, was essential for mediating its interaction with PD-L1. The mAb STM418 specifically targeted glycosylated PD-1, exhibiting higher binding affinity to PD-1 than FDA-approved PD-1 antibodies, potently inhibiting PD-L1/PD-1 binding, and enhancing antitumor immunity. Together, these findings provide novel insights into the functional significance of PD-1 glycosylation and offer a rationale for targeting glycosylated PD-1 as a potential strategy for immunotherapy. SIGNIFICANCE: These findings demonstrate that glycosylation of PD-1 is functionally significant and targeting glycosylated PD-1 may serve as a means to improve immunotherapy response.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/metabolism , Female , Glycosylation , HEK293 Cells , Heterografts , Humans , Jurkat Cells , Leukemia, T-Cell/metabolism , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Nivolumab/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
AAPS J ; 22(2): 22, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31900688

ABSTRACT

Immuno-PET is a molecular imaging technique utilizing positron emission tomography (PET) to measure the biodistribution of an antibody species labeled with a radioactive isotope. When applied as a clinical imaging technique, an immuno-PET imaging agent must be manufactured with quality standards appropriate for regulatory approval. This paper describes methods relevant to the chemistry, manufacturing, and controls component of an immuno-PET regulatory filing, such as an investigational new drug application. Namely, the production, quality control, and characterization of the immuno-PET clinical imaging agent, ZED8, an 89Zr-labeled CD8-specific monovalent antibody as well as its desferrioxamine-conjugated precursor, CED8, is described and evaluated. PET imaging data in a human CD8-expressing tumor murine model is presented as a proof of concept that the imaging agent exhibits target specificity and comparable biodistribution across a range of desferrioxamine conjugate loads.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Leukemia, T-Cell/diagnostic imaging , Molecular Imaging , Positron-Emission Tomography , Radioisotopes/administration & dosage , Radiopharmaceuticals/administration & dosage , Zirconium/administration & dosage , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Female , Humans , Leukemia, T-Cell/immunology , Mice, SCID , Predictive Value of Tests , Proof of Concept Study , Quality Control , Radioisotopes/chemistry , Radioisotopes/standards , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/standards , Zirconium/chemistry , Zirconium/standards
8.
Cytotherapy ; 21(7): 769-781, 2019 07.
Article in English | MEDLINE | ID: mdl-31160157

ABSTRACT

Immunotherapy with chimeric antigen receptor T (CAR-T) cells has proved remarkably effective in recently published clinical trials. In this meta-analysis, we performed a systematic review in terms of the clinical response treated with CAR-T cells in acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL) and lymphomas patients. Thirty-eight published clinical studies including 665 patients were eligible for response rate (RR) evaluation. The overall pooled RR of CD19-CAR-T cells was 72% (95% confidence interval: 62-77%). The various clinical parameters were analyzed. RR was 81% in ALL, 68% in lymphoma and 70% in CLL. RR in patients who received interleukin (IL)-2 was 70%, whereas in those who did not receive IL-2, it was 74%. RR was 75% with lymphodepletion and 56% without lymphodepletion. RR with autologous cells was 76% and 57% with allogeneic cells. In conclusion, this meta-analysis showed a high clinical RR of CD19-CAR-T cell-based immunotherapy in patients with refractory B-cell malignancies.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/immunology , Leukemia, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Clinical Trials as Topic , Female , Humans , Immunotherapy, Adoptive/adverse effects , Leukemia, B-Cell/therapy , Leukemia, T-Cell/therapy , Lymphoma/immunology , Lymphoma/therapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/therapy , Male , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
9.
Leukemia ; 32(9): 1970-1983, 2018 09.
Article in English | MEDLINE | ID: mdl-29483708

ABSTRACT

T cell malignancies represent a group of hematologic cancers with high rates of relapse and mortality in patients for whom no effective targeted therapies exist. The shared expression of target antigens between chimeric antigen receptor (CAR) T cells and malignant T cells has limited the development of CAR-T because of unintended CAR-T fratricide and an inability to harvest sufficient autologous T cells. Here, we describe a fratricide-resistant "off-the-shelf" CAR-T (or UCART7) that targets CD7+ T cell malignancies and, through CRISPR/Cas9 gene editing, lacks both CD7 and T cell receptor alpha chain (TRAC) expression. UCART7 demonstrates efficacy against human T cell acute lymphoblastic leukemia (T-ALL) cell lines and primary T-ALL in vitro and in vivo without the induction of xenogeneic GvHD. Fratricide-resistant, allo-tolerant "off-the-shelf" CAR-T represents a strategy for treatment of relapsed and refractory T-ALL and non-Hodgkin's T cell lymphoma without a requirement for autologous T cells.


Subject(s)
Immunotherapy, Adoptive , Leukemia, T-Cell/immunology , Leukemia, T-Cell/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Animals , Antigens, CD7/genetics , Antigens, CD7/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CRISPR-Cas Systems , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Gene Deletion , Gene Editing , Gene Order , Genetic Vectors/genetics , Humans , Immunotherapy, Adoptive/methods , Leukemia, T-Cell/genetics , Leukemia, T-Cell/therapy , Male , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
10.
Immunobiology ; 223(1): 73-80, 2018 01.
Article in English | MEDLINE | ID: mdl-29030004

ABSTRACT

Tumor-associated macrophages (TAMs) are well accepted and the pathological role of macrophages in hematopoietic malignancies have been proposed. Hepatomegaly is frequently observed in T cell acute lymphoblastic leukemia (T-ALL) patients with poor prognosis. However, the role of leukemia-associated macrophages (LAMs) in hepatic microenvironment remains unclear. Here, the characteristics of hepatic LAMs (H-LAMs) were studied in Notch1 induced T-ALL model. Increase in proportion and absolute counts of H-LAMs was detected with infiltration of inflammatory cells. Furthermore, H-LAMs exhibited a more M1-like phenotype distinct from that of TAMs in hepatocellular carcinoma and LAMs from BM or spleen in leukemia. Moreover, H-LAMs expressed increased level of cytokines in charge of recruiting inflammatory cells, which contributed to pro-inflammatory hepatic microenvironment.


Subject(s)
Inflammation/immunology , Leukemia, T-Cell/immunology , Liver Neoplasms/immunology , Macrophages/immunology , Th1 Cells/immunology , Animals , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Phenotype , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Tumor Microenvironment
11.
Nat Med ; 23(12): 1416-1423, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29131157

ABSTRACT

Mature T cell cancers are typically aggressive, treatment resistant and associated with poor prognosis. Clinical application of immunotherapeutic approaches has been limited by a lack of target antigens that discriminate malignant from healthy (normal) T cells. Unlike B cell depletion, pan-T cell aplasia is prohibitively toxic. We report a new targeting strategy based on the mutually exclusive expression of T cell receptor ß-chain constant domains 1 and 2 (TRBC1 and TRBC2). We identify an antibody with unique TRBC1 specificity and use it to demonstrate that normal and virus-specific T cell populations contain both TRBC1+ and TRBC2+ compartments, whereas malignancies are restricted to only one. As proof of concept for anti-TRBC immunotherapy, we developed anti-TRBC1 chimeric antigen receptor (CAR) T cells, which recognized and killed normal and malignant TRBC1+, but not TRBC2+, T cells in vitro and in a disseminated mouse model of leukemia. Unlike nonselective approaches targeting the entire T cell population, TRBC-targeted immunotherapy could eradicate a T cell malignancy while preserving sufficient normal T cells to maintain cellular immunity.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy, Adoptive/methods , Leukemia, T-Cell/therapy , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Jurkat Cells , K562 Cells , Leukemia, T-Cell/immunology , Male , Mice , Molecular Targeted Therapy/methods , T-Lymphocytes/immunology
12.
Nat Commun ; 8(1): 427, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874664

ABSTRACT

Mammalian cells use cytosolic nucleic acid receptors to detect pathogens and other stress signals. In innate immune cells the presence of cytosolic DNA is sensed by the cGAS-STING signalling pathway, which initiates a gene expression programme linked to cellular activation and cytokine production. Whether the outcome of the STING response varies between distinct cell types remains largely unknown. Here we show that T cells exhibit an intensified STING response, which leads to the expression of a distinct set of genes and results in the induction of apoptosis. Of note, this proapoptotic STING response is still functional in cancerous T cells and delivery of small molecule STING agonists prevents in vivo growth of T-cell-derived tumours independent of its adjuvant activity. Our results demonstrate how the magnitude of STING signalling can shape distinct effector responses, which may permit for cell type-adjusted behaviours towards endogenous or exogenous insults.The cGAS/STING signalling pathway is responsible for sensing intracellular DNA and activating downstream inflammatory genes. Here the authors show mouse primary T cells and T leukaemia are hyperresponsive to STING agonist, and this strong STING signalling is associated with apoptosis induction.


Subject(s)
Apoptosis , Membrane Proteins/metabolism , Signal Transduction , Animals , Interferon Regulatory Factor-3/metabolism , Leukemia, T-Cell/immunology , Leukemia, T-Cell/pathology , Mice, Inbred C57BL , Protein Binding , T-Lymphocytes/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
13.
Am J Hematol ; 92(9): 892-901, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28543380

ABSTRACT

With the emerging success of treating CD19 expressing B cell malignancies with ex vivo modified, autologous T cells that express CD19-directed chimeric antigen receptors (CAR), there is intense interest in expanding this evolving technology to develop effective modalities to treat other malignancies including solid tumors. Exploiting this approach to develop a therapeutic modality for T cell malignancies for which the available regimens are neither curative, nor confer long term survival we generated a lentivirus-based CAR gene transfer system to target the chemokine receptor CCR4 that is over-expressed in a spectrum of T cell malignancies as well as in CD4+ CD25+ Foxp3+ T regulatory cells that accumulate in the tumor microenvironment constituting a barrier against anti-tumor immunity. Ex vivo modified, donor-derived T cells that expressed CCR4 directed CAR displayed antigen-dependent potent cytotoxicity against patient-derived cell lines representing ATL, CTCL, ALCL and a subset of HDL. Furthermore, these CAR T cells also eradicated leukemia in a mouse xenograft model of ATL illustrating the potential utility of this modality in the treatment of a wide spectrum of T cell malignancies.


Subject(s)
Hematologic Neoplasms , Receptors, Antigen, T-Cell , Receptors, CCR4/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/immunology , Leukemia, T-Cell/pathology , Leukemia, T-Cell/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, CCR4/genetics , Receptors, CCR4/immunology , T-Lymphocytes, Regulatory/pathology , Xenograft Model Antitumor Assays
14.
Exp Oncol ; 39(1): 36-41, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28361854

ABSTRACT

The aim of this study was to characterize the proliferative activity of the anti-histone H1 IgGs towards human T-leukaemia CEM cells. MATERIALS AND METHODS: Anti-histone H1 IgGs were purified from blood serum of systemic lupus erythematosus patients by precipitation of serum proteins with 50% ammonium sulfate followed by a sequential affinity chromatography on Protein G-Sepharose and histone H1-Sepharose columns. To avoid contamination with other proteins, anti-histone H1 IgGs were subjected to strongly acidic pH 2.0 during gel filtration through HPLC column. The effects of the anti-histone H1 IgGs on cell viability and cell cycle were tested by MTS-assay and flow cytometry, correspondingly. The cross-reactivity of the anti-histone H1 antibodies towards heterogenetic and cellular antigens was evaluated by Western-blot analysis. RESULTS: It was found that incubation of CEM cells with the HPLC-purified anti-histone H1 IgGs resulted in significant stimulation of cell growth by 46% after 48 h of incubation. These IgGs possess an antigenic poly-specificity to positively charged heterogenetic antigens and different cellular antigens. FITC-labeled and biotinylated anti-histone H1 IgGs are internalized by CEM cells and preferentially accumulated in the cytoplasm. CONCLUSION: The anti-histone H1 IgGs are shown to internalize human T-leukemia CEM and stimulate their proliferation. These IgGs are polyspecific toward cellular antigens.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Histones/immunology , Antibodies, Anti-Idiotypic/isolation & purification , Antibodies, Anti-Idiotypic/metabolism , Antibody Affinity/immunology , Antibody Specificity/immunology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, Affinity , Chromatography, Gel , Cross Reactions/immunology , Cytoplasm/metabolism , Humans , Leukemia, T-Cell/immunology , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology
15.
Oncotarget ; 8(15): 24706-24718, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28160557

ABSTRACT

Many human cancers have been reported to have enhanced expression of the immune checkpoint molecule programmed death-ligand 1 (PD-L1), which binds to programmed cell death-1 (PD-1) expressed on immune cells. PD-L1/PD-1 plays a role in inhibition of antitumor immunity by inducing T cell apoptosis and tolerance. Thus, it is crucial to elucidate mechanisms of PD-L1 expression on cancer cells. ERO1-α is an oxidase located in the endoplasmic reticulum. It is overexpressed in a variety of tumor types and it plays a role in disulfide bond formation in collaboration with PDI. Here, we investigated the influence of ERO1-α on expression of PD-L1 and immune escape. We demonstrated that ERO1-α augmented the expression of PD-L1 via facilitation of oxidative protein folding within PD-L1. In addition, we showed that overexpression of ERO1-α increased HIF-1α protein expression, resulting in an increase of PD-L1 mRNA as well as protein. In clinical cases, we observed that the expression of ERO1-α in triple negative breast cancer was related to the expression of PD-L1. Moreover, apoptosis of Jurkat leukemia T cells, which express PD-1, induced by tumor PD-L1 was inhibited when ERO1-α was depleted. The results suggest that targeting ERO1-α in tumor cells can be a novel approach for cancer immunotherapy. Therefore, the role of ERO1-α in tumor-mediated immunosuppression should be further explored.


Subject(s)
B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Leukemia, T-Cell/immunology , Membrane Glycoproteins/immunology , Oxidoreductases/immunology , Apoptosis/immunology , B7-H1 Antigen/biosynthesis , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Interferon-gamma/pharmacology , Jurkat Cells , Leukemia, T-Cell/enzymology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/biosynthesis , Protein Folding , Transfection , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/immunology , Tumor Escape , Up-Regulation/drug effects , Up-Regulation/immunology
16.
Cell Immunol ; 313: 59-66, 2017 03.
Article in English | MEDLINE | ID: mdl-28087047

ABSTRACT

LMB-2, is a potent recombinant immunotoxin (RIT) that is composed of scFv antibody that targets CD25 (Tac) and a toxin fragment (PE38). It is used to treat T cell leukemias and lymphomas. To make LMB-2 less immunogenic, we introduced a large deletion in domain II and six point mutations in domain III that were previously shown to reduce T cell activation in other RITs. We found that unlike other RITs, deletion of domain II from LMB-2 severely compromised its activity. Rather than deletion, we identified T cell epitopes in domain II and used alanine substitutions to identify point mutations that diminished those epitopes. The novel RIT, LMB-142 contains a 38kDa toxin and nine point mutations that diminished T cell response to the corresponding peptides by an average of 75%. LMB-142 has good cytotoxic activity and has lower nonspecific toxicity in mice. LMB-142 should be more efficient in cancer therapy because more treatment cycles can be given.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Immunotoxins/therapeutic use , Leukemia, T-Cell/therapy , Pseudomonas/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/genetics , Bacterial Toxins/genetics , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Drug Design , Enzyme-Linked Immunospot Assay , Epitopes, T-Lymphocyte/genetics , Exotoxins/genetics , Exotoxins/therapeutic use , Female , Genetic Engineering , Humans , Immunotoxins/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Leukemia, T-Cell/immunology , Lymphocyte Activation , Mice , Mutagenesis, Site-Directed , Mutation/genetics
17.
Nat Med ; 23(1): 18-27, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28060797

ABSTRACT

T memory stem (TSCM) cells are a rare subset of memory lymphocytes endowed with the stem cell-like ability to self-renew and the multipotent capacity to reconstitute the entire spectrum of memory and effector T cell subsets. Cumulative evidence in mice, nonhuman primates and humans indicates that TSCM cells are minimally differentiated cells at the apex of the hierarchical system of memory T lymphocytes. Here we describe emerging findings demonstrating that TSCM cells, owing to their extreme longevity and robust potential for immune reconstitution, are central players in many physiological and pathological human processes. We also discuss how TSCM cell stemness could be leveraged therapeutically to enhance the efficacy of vaccines and adoptive T cell therapies for cancer and infectious diseases or, conversely, how it could be disrupted to treat TSCM cell driven and sustained diseases, such as autoimmunity, adult T cell leukemia and HIV-1.


Subject(s)
Cell Self Renewal/immunology , Immunologic Memory/immunology , Stem Cells/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Cell Differentiation , HIV Infections/immunology , HIV Infections/therapy , Humans , Leukemia, T-Cell/immunology , Leukemia, T-Cell/therapy , T-Lymphocytes/transplantation
18.
Cancer Discov ; 6(9): 972-85, 2016 09.
Article in English | MEDLINE | ID: mdl-27354269

ABSTRACT

UNLABELLED: Cancer onset and progression involves the accumulation of multiple oncogenic hits, which are thought to dominate or bypass the physiologic regulatory mechanisms in tissue development and homeostasis. We demonstrate in T-cell acute lymphoblastic leukemia (T-ALL) that, irrespective of the complex oncogenic abnormalities underlying tumor progression, experimentally induced, persistent T-cell receptor (TCR) signaling has antileukemic properties and enforces a molecular program resembling thymic negative selection, a major developmental event in normal T-cell development. Using mouse models of T-ALL, we show that induction of TCR signaling by high-affinity self-peptide/MHC or treatment with monoclonal antibodies to the CD3ε chain (anti-CD3) causes massive leukemic cell death. Importantly, anti-CD3 treatment hampered leukemogenesis in mice transplanted with either mouse- or patient-derived T-ALLs. These data provide a strong rationale for targeted therapy based on anti-CD3 treatment of patients with TCR-expressing T-ALL and demonstrate that endogenous developmental checkpoint pathways are amenable to therapeutic intervention in cancer cells. SIGNIFICANCE: T-ALLs are aggressive malignant lymphoid proliferations of T-cell precursors characterized by high relapse rates and poor prognosis, calling for the search for novel therapeutic options. Here, we report that the lineage-specific TCR/CD3 developmental checkpoint controlling cell death in normal T-cell progenitors remains switchable to induce massive tumor cell apoptosis in T-ALL and is amenable to preclinical therapeutic intervention. Cancer Discov; 6(9); 972-85. ©2016 AACR.See related commentary by Lemonnier and Mak, p. 946This article is highlighted in the In This Issue feature, p. 932.


Subject(s)
Leukemia, T-Cell/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Apoptosis/drug effects , Apoptosis/immunology , CD3 Complex/immunology , CD3 Complex/metabolism , Clonal Selection, Antigen-Mediated , Disease Models, Animal , Female , Humans , Immunophenotyping , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/genetics , Leukemia, T-Cell/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Signal Transduction/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
19.
Oncotarget ; 7(16): 21199-221, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27028870

ABSTRACT

Immunotherapy of cancer envisions the adoptive transfer of T-cells genetically engineered with tumor-specific heterodimeric α/ß T-cell receptors (TCRα/ß). However, potential mispairing of introduced TCRα/ß-chains with endogenous ß/α-ones may evoke unpredictable autoimmune reactivities. A novel single chain (sc)TCR format relies on the fusion of the Vα-Linker-Vß-fragment to the TCR Cß-domain and coexpression of the TCR Cα-domain capable of recruiting the natural CD3-complex for full and hence, native T-cell signaling. Here, we tested whether such a gp100(280-288)- or p53(264-272) tumor antigen-specific scTCR is still prone to mispairing with TCRα. In a human Jurkat-76 T-cell line lacking endogenous TCRs, surface expression and function of a scTCR could be reconstituted by any cointroduced TCRα-chain indicating mispairing to take place on a molecular basis. In contrast, transduction into human TCRα/ß-positive T-cells revealed that mispairing is largely reduced. Competition experiments in Jurkat-76 confirmed the preference of dcTCR to selfpair and to spare scTCR. This also allowed for the generation of dc/scTCR-modified cytomegalovirus/tumor antigen-bispecific T-cells to augment T-cell activation in CMV-infected tumor patients. Residual mispairing was prevented by strenghtening the Vα-Li-Vß-fragment through the design of a novel disulfide bond between a Vα- and a linker-resident residue close to Vß. Multimer-stainings, and cytotoxicity-, IFNγ-secretion-, and CFSE-proliferation-assays, the latter towards dendritic cells endogenously processing RNA-electroporated gp100 antigen proved the absence of hybrid scTCR/TCRα-formation without impairing avidity of scTCR/Cα in T-cells. Moreover, a fragile cytomegalovirus pp65(495-503)-specific scTCR modified this way acquired enhanced cytotoxicity. Thus, optimized scTCR/Cα inhibits residual TCR mispairing to accomplish safe adoptive immunotherapy for bulk endogenous TCRα/ß-positive T-cells.


Subject(s)
CD3 Complex/immunology , Leukemia, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Biomarkers, Tumor , CD3 Complex/genetics , Cell Membrane , Cell Proliferation , Humans , Immunotherapy, Adoptive , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Mice , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signal Transduction , T-Lymphocytes/metabolism , Tumor Cells, Cultured
20.
Transplantation ; 100(1): 217-26, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26285015

ABSTRACT

BACKGROUND: Assessing the serum reactivity to HLA is essential for the evaluation of transplant candidates and the follow-up of allograft recipients. In this study, we look for evidence at the clonal level that polyreactive antibodies cross-reactive to apoptotic cells and multiple autoantigens can also react to HLA and contribute to the overall serum reactivity. METHODS: We immortalized B cell clones from the blood of 2 kidney transplant recipients and characterized their reactivity to self-antigens, apoptotic cells as well as native, denatured, and cryptic HLA determinants using enzyme-linked immunosorbent assay (ELISA), immunofluorescence, flow cytometry and Luminex assays. We also assessed the reactivity of 300 pretransplant serum specimens to HLA and apoptotic cells. RESULTS: We report here 4 distinct B cell clones cross-reactive to self and HLA class I. All 4 clones reacted to numerous HLA class I alleles but did not appear to target canonical "shared" epitopes. In parallel experiments, we observed a strong correlation between IgG reactivity to HLA and apoptotic cells in pretransplant serum samples collected from 300 kidney transplant recipients. Further analysis revealed that samples with higher reactivity to apoptotic cells displayed significantly higher class I percent panel-reactive antibodies compared to samples with low reactivity to apoptotic cells. CONCLUSIONS: We provide here (1) proof of principle at the clonal level that human polyreactive antibodies can cross-react to HLA, multiple self-antigens and apoptotic cells and (2) supportive evidence that polyreactive antibodies contribute to overall HLA reactivity in the serum of patients awaiting kidney transplant.


Subject(s)
B-Lymphocytes/immunology , HLA Antigens/immunology , Histocompatibility , Isoantibodies/blood , Kidney Transplantation , Transplant Recipients , Apoptosis , Autoantigens , Boston , Clone Cells , Coculture Techniques , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitopes , Feeder Cells , Flow Cytometry , Fluorescent Antibody Technique , Genes, Immunoglobulin Heavy Chain , HEK293 Cells , Histocompatibility Testing , Humans , Immunoglobulin Variable Region/genetics , Jurkat Cells , Leukemia, T-Cell/immunology , Leukemia, T-Cell/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...