Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 5387, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31772172

ABSTRACT

T cell-engaging immunotherapies are changing the landscape of current cancer care. However, suitable target antigens are scarce, restricting these strategies to very few tumor types. Here, we report on a T cell-engaging antibody derivative that comes in two complementary halves and addresses antigen combinations instead of single molecules. Each half, now coined hemibody, contains an antigen-specific single-chain variable fragment (scFv) fused to either the variable light (VL) or variable heavy (VH) chain domain of an anti-CD3 antibody. When the two hemibodies simultaneously bind their respective antigens on a single cell, they align and reconstitute the original CD3-binding site to engage T cells. Employing preclinical models for aggressive leukemia and breast cancer, we show that by the combinatorial nature of this approach, T lymphocytes exclusively eliminate dual antigen-positive cells while sparing single positive bystanders. This allows for precision targeting of cancers not amenable to current immunotherapies.


Subject(s)
Antibodies/pharmacology , Antineoplastic Agents, Immunological/pharmacology , CD3 Complex/metabolism , Immunotherapy/methods , T-Lymphocytes/immunology , Animals , Antibodies/genetics , Antineoplastic Agents, Immunological/immunology , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Bystander Effect , Cell Line, Tumor , Female , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Lymphocyte Activation , Mice, Inbred BALB C , Mice, Inbred NOD , Precision Medicine/methods , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Xenograft Model Antitumor Assays
2.
Mol Ther ; 21(11): 2122-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23831595

ABSTRACT

In a phase I study of autologous chimeric antigen receptor (CAR) anti-LeY T-cell therapy of acute myeloid leukemia (AML), we examined the safety and postinfusion persistence of adoptively transferred T cells. Following fludarabine-containing preconditioning, four patients received up to 1.3 × 109 total T cells, of which 14-38% expressed the CAR. Grade 3 or 4 toxicity was not observed. One patient achieved a cytogenetic remission whereas another with active leukemia had a reduction in peripheral blood (PB) blasts and a third showed a protracted remission. Using an aliquot of In111-labeled CAR T cells, we demonstrated trafficking to the bone marrow (BM) in those patients with the greatest clinical benefit. Furthermore, in a patient with leukemia cutis, CAR T cells infiltrated proven sites of disease. Serial PCR of PB and BM for the LeY transgene demonstrated that infused CAR T cells persisted for up to 10 months. Our study supports the feasibility and safety of CAR-T-cell therapy in high-risk AML, and demonstrates durable in vivo persistence.


Subject(s)
Immunotherapy, Adoptive , Leukemia, Myeloid, Acute/therapy , Lewis Blood Group Antigens/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Aged , Bone Marrow/immunology , Female , Humans , Leukemia, Myeloid, Acute/immunology , Male , Middle Aged , Remission Induction , Transplantation Conditioning , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
5.
J Immunother ; 32(3): 292-301, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19242371

ABSTRACT

In this study we aimed to determine the suitability of the Lewis-Y carbohydrate antigen as a target for immunotherapy using genetically redirected T cells. Using the 3S193 monoclonal antibody and immunohistochemistry, Lewis-Y was found to be expressed on a range of tumors including 42% squamous cell lung carcinoma, 80% lung adenocarcinoma, 25% ovarian carcinoma, and 25% colorectal adenocarcinoma. Expression levels varied from low to intense on between 1% and 90% of tumor cells. Lewis- was also found in soluble form in sera from both normal donors and cancer patients using a newly developed enzyme-linked immunosorbent assay. Serum levels in patients was often less than 1 ng/mL, similar to normal donors, but approximately 30% of patients had soluble Lewis-Y levels exceeding 1 ng/mL and up to 9 ng/mL. Lewis-Y-specific human T cells were generated by genetic modification with a chimeric receptor encoding a single-chain humanized antibody linked to the T-cell signaling molecules, T-cell receptor-zeta, and CD28. T cells responded against the Lewis-Y antigen by cytokine secretion and cytolysis in response to tumor cells. Importantly, the T-cell response was not inhibited by patient serum containing soluble Lewis-Y. This study demonstrates that Lewis-Y is expressed on a large number of tumors and Lewis-Y-specific T cells can retain antitumor function in the presence of patient serum, indicating that this antigen is a suitable target for this form of therapy.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy, Adoptive , Lewis Blood Group Antigens/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Antigens, Neoplasm/blood , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lewis Blood Group Antigens/blood , Lewis Blood Group Antigens/genetics , Neoplasms/pathology , T-Lymphocytes/immunology , Transduction, Genetic
6.
J Immunother ; 31(2): 166-79, 2008.
Article in English | MEDLINE | ID: mdl-18481386

ABSTRACT

Dendritic cell (DC) immunotherapy is being actively studied in multiple myeloma (MM). We aimed to use positron emission tomography or single positron emission tomography to determine the in vivo distribution of monocyte-derived nonmatured DC or matured DC (mDC) administered to patients with MM. Eligible patients had stable or slowly progressive MM and elevated serum MUC-1 or MUC-1 expression on marrow plasma cells. DCs were derived from granulocyte-macrophage colony-stimulating factor+ interleukin-13 stimulated autologous monocytes, pulsed with mannan-MUC1 fusion protein, and matured by FMKp and interferon-gamma. Before injection, DCs were labeled with either 18fluorine-fluorodeoxyglucose, 111indium-oxine or 64copper-pyruvaldehyde-bis-N-4-methylthiosemicarbazone. Labeled DCs were given either as a single intravenous dose or by concurrent subcutaneous (SC), intradermal (ID), and intranodal routes. 18Fluorine-fluorodeoxyglucose tracking was unsuccessful owing to high radiolabel efflux. 64Copper-pyruvaldehyde-bis-N-4-methylthiosemicarbazone-labeled mDC (n=2 patients) demonstrated tracking to regional nodes but quantitation was also limited owing to cellular efflux. 111Indium-oxine, however, gave reproducible tracking of both nmDc and mDC (n=6) to regional lymph node after either SC or ID administration, with mDC revealing superior migration to regional lymph node. SC and ID routes produced similar levels of DC migration.


Subject(s)
Cell Movement/immunology , Dendritic Cells/transplantation , Multiple Myeloma/therapy , Aged , Antibodies/blood , Antibodies/immunology , Antigens, Bacterial/pharmacology , Antigens, CD/analysis , Cytokines/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Female , Fluorodeoxyglucose F18/chemistry , Humans , Interferon-gamma/metabolism , Klebsiella pneumoniae/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Lymph Nodes/pathology , Male , Middle Aged , Mucin-1/blood , Mucin-1/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Organometallic Compounds/chemistry , Oxyquinoline/analogs & derivatives , Oxyquinoline/chemistry , Radiopharmaceuticals/chemistry , Spleen/pathology , Thiosemicarbazones/chemistry , Tomography, Emission-Computed/methods
7.
Haematologica ; 92(8): 1075-82, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17640854

ABSTRACT

BACKGROUND AND OBJECTIVES: In vitro studies suggest that thalidomide has an immunoregulatory role and alters the marrow microenvironment. We assessed laboratory and clinical parameters in patients with myeloma treated with thalidomide as potential prognostic markers and looked for changes with therapy. DESIGN AND METHODS: Seventy-five patients with relapsed/refractory myeloma received thalidomide in a phase II trial. Serial samples of platelet-poor plasma and bone marrow were tested for angiogenic cytokines including vascular endothelial growth factor (VEGF), marrow microvessel-density (MVD), mast cells and CD57+ cell expression. The effects of these parameters on response rate (RR), progression-free survival (PFS) and overall survival (OS) were analyzed. RESULTS: Elevated baseline VEGF predicted for a superior RR (p=0.018) and PFS. Elevated CD57+ cells also predicted superior PFS (p=0.012). MVD did not predict for RR, PFS or OS, but MVD and VEGF fell significantly in responders. Multivariate analysis identified that inferior OS was associated with age >65 years (p=0.017), raised lactate dehydrogenase (p=0.001), raised hepatocyte growth factor levels (p=0.012) and low pre-treatment CD57+ cells (p<0.001). INTERPRETATION AND CONCLUSIONS: Our findings support the suggestion that thalidomide has anti-angiogenic and immunomodulatory effects in myeloma. The preferred method for assessing angiogenesis is plasma VEGF levels and the assessment of CD57+ cells for patients with myeloma receiving novel immunomodulatory drugs should be further investigated.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Bone Marrow/pathology , CD57 Antigens/analysis , Cytokines/blood , Immunologic Factors/therapeutic use , Mast Cells/pathology , Multiple Myeloma/drug therapy , T-Lymphocytes, Cytotoxic/pathology , Thalidomide/therapeutic use , Adult , Aged , Clinical Trials, Phase II as Topic/statistics & numerical data , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multicenter Studies as Topic/statistics & numerical data , Multiple Myeloma/blood , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Neoplasm Proteins/blood , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Prognosis , Survival Analysis , T-Lymphocytes, Cytotoxic/chemistry , Treatment Outcome
11.
Proc Natl Acad Sci U S A ; 102(52): 19051-6, 2005 Dec 27.
Article in English | MEDLINE | ID: mdl-16365285

ABSTRACT

In this study, human T cells were provided with a reactivity against the Lewis-Y (Le(Y)) carbohydrate antigen, which is overexpressed on 70% of epithelial-derived tumors, but not normally recognized by T cells. Antitumor reactivity was achieved by transduction of T cells with a gene encoding a cell-surface chimeric receptor composed of single-chain anti-Le(Y) antibody linked to an enhanced cytoplasmic signaling domain made up of CD28 and CD3-zeta. Importantly, the single-chain antibody was humanized to try to reduce potential problems of human anti-mouse antibody responses in patients receiving chimeric receptor-modified T cells in future clinical trials. T cells expressing the chimeric receptor were demonstrated to secrete cytokines and proliferate in response to receptor ligation and lysed Le(Y+) tumors in vitro. Another aspect of this study was the finding that no activity was observed against normal tissue, as represented by autologous neutrophils that express low levels of Le(Y). Significantly, systemic delivery of anti-Le(Y) T cells dramatically inhibited established s.c. human ovarian OVCAR-3 tumors (a recognized difficult model to treat) in mice. Finally, we demonstrated that anti-Le(Y) T cells preferentially expanded or accumulated in the tumor compared with control empty vector T cells, thereby providing mechanistic insight into the specific antitumor response. This study supports the use of humanized gene-modified T cells as a potential therapy for Le(Y+) malignancies.


Subject(s)
Adoptive Transfer , Immunotherapy/methods , Lewis Blood Group Antigens/chemistry , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/cytology , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , Autoimmunity , CD28 Antigens/biosynthesis , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Clinical Trials as Topic , Cytokines/metabolism , Flow Cytometry , Genetic Vectors , Humans , Immunohistochemistry , Leukocytes, Mononuclear/cytology , Mice , Neoplasm Transplantation , Neoplasms/metabolism , Neutrophils/metabolism , Recombinant Fusion Proteins/chemistry , Retroviridae/genetics , Sialic Acid Binding Ig-like Lectin 3 , T-Lymphocytes/metabolism , Time Factors
13.
Ann Hematol ; 84(2): 118-21, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15452669

ABSTRACT

Enteropathy-associated T-cell lymphoma (EATL) ultimately develops in 7-10% of patients with long-standing coeliac disease. In patients without a prior diagnosis of coeliac disease this is a very rare disorder, and the diagnosis in such cases is often difficult and delayed due to the non-specific nature of the symptoms and a very low index of clinical suspicion. Standard anti-lymphoma therapies have minimal utility in patients with EATL, and their prognosis is poor. An added difficulty is the high risk of intestinal perforation especially with the commencement of treatment due to the multifocal nature of bowel disease and poor underlying nutrition and tissue integrity. To illustrate these problems and provide an example of how these issues may be addressed, we report the case of a patient with EATL who was completely asymptomatic from unsuspected underlying coeliac disease and presented initially with back pain followed by bowel obstruction. He was treated with gut rest with total parenteral nutrition before commencing an intensive chemotherapy regimen [hyper-CVAD (cyclophosphamide, vincristine, doxorubicin, and dexamethasone)] and is currently well in ongoing complete remission 34 months later.


Subject(s)
Lymphoma, T-Cell/complications , Malabsorption Syndromes/diagnosis , Adult , Back Pain , Celiac Disease , Diagnosis, Differential , Disease Management , Humans , Intestinal Obstruction , Lymphoma, T-Cell/therapy , Malabsorption Syndromes/etiology , Malabsorption Syndromes/therapy , Male , Remission Induction
14.
J Leukoc Biol ; 75(6): 1022-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15020647

ABSTRACT

Despite the important role immunoglobulin G (IgG)-secreting plasma cells play in memory immune responses, the differentiation and homeostasis of these cells are not completely understood. Here, we studied the differentiation of human IgG-secreting cells ex vivo and in vitro, identifying these cells by the cellular affinity matrix technology. Several subpopulations of IgG-secreting cells were identified among the cells isolated from tonsils and bone marrow, particularly differing in the expression levels of CD9, CD19, and CD38. CD38 low IgG-secreting cells were present exclusively in the tonsils. A major fraction of these cells appeared to be early plasma cell precursors, as upon activation of B cells in vitro, IgG secretion preceded up-regulation of CD38, and on tonsillar sections, IgG-containing, CD38 low cells with a plasmacytoid phenotype were found in follicles, where plasma cell differentiation starts. A unitary phenotype of migratory peripheral blood IgG-secreting cells suggests that all bone marrow plasma cell populations share a common precursor cell. These data are compatible with a multistep model for plasma cell differentiation and imply that a common CD38 low IgG-secreting precursor gives rise to a diverse plasma cell compartment.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Antigens, CD/metabolism , Bone Marrow/immunology , Immunoglobulin G/metabolism , Palatine Tonsil/immunology , Plasma Cells/immunology , ADP-ribosyl Cyclase 1 , Antigens, CD19/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Differentiation , Cell Movement , Humans , In Vitro Techniques , Membrane Glycoproteins/metabolism , Palatine Tonsil/cytology , Phenotype , Plasma Cells/metabolism , Tetraspanin 29
15.
Blood ; 100(9): 3311-8, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12384432

ABSTRACT

The interleukin 6/glycoprotein 130/signal transducer and activator of transcription 3 (IL-6/gp130/STAT3) pathway has been reported to play an important role in the pathogenesis of multiple myeloma (MM) and for survival of MM cells. However, most data concerning the role of IL-6 and IL-6-triggered signaling pathways were obtained from experiments performed with MM cell lines and without considering the bone marrow microenvironment. Thus, the precise role of IL-6 and its intracellular signaling pathways for survival of human MM cells is still unclear. Here we show that treatment of human MM cells (IL-6-dependent MM cell line INA-6 and primary MM cells) with the IL-6 receptor antagonist Sant7 or with an anti-gp130 monoclonal antibody (mAb) induced apoptosis if the cells were cultured in the absence of bone marrow stromal cells (BMSCs). In contrast, apoptosis could not be observed if the MM cells were cocultured with BMSCs. The analysis of intracellular pathways revealed that Sant7 and anti-gp130 mAb were effectively inhibiting the phosphorylation of gp130 and STAT3 in the absence and presence of BMSCs, whereas ERK1 and ERK2 (ERK1,2) phosphorylation was only slightly affected. In contrast, treatment with the farnesyl transferase inhibitor, FPT III, induced apoptosis in MM cells in the absence or presence of BMSCs and led to a complete inhibition of the Ras/mitogen-activated protein kinase pathway. These observations indicate that the IL-6/gp130/STAT3 pathway is not essential for survival of human myeloma cells if they are grown in the presence of cells from the bone marrow microenvironment. Furthermore, we provide evidence that farnesyl transferase inhibitors might be useful for the development of novel therapeutic strategies for the treatment of MM.


Subject(s)
Bone Marrow Cells/physiology , Interleukin-6/analogs & derivatives , Multiple Myeloma/pathology , Neoplastic Stem Cells/cytology , Signal Transduction/physiology , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/physiology , Antibodies, Monoclonal/pharmacology , Antigens, CD/physiology , Apoptosis , Cell Survival , Coculture Techniques , Cytokine Receptor gp130 , DNA-Binding Proteins/physiology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , Humans , Interleukin-6/pharmacology , Interleukin-6/physiology , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Organophosphonates/pharmacology , Phosphorylation , Protein Prenylation , Protein Processing, Post-Translational , Receptors, Interleukin-6/antagonists & inhibitors , STAT3 Transcription Factor , Signal Transduction/drug effects , Stromal Cells/physiology , Trans-Activators/physiology , Tumor Cells, Cultured/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...