Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunol Cell Biol ; 93(2): 198-212, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25385067

ABSTRACT

Interleukin-17A (IL-17A) is a pro-inflammatory cytokine that has an important role at mucosal sites in a wide range of immune responses including infection, allergy and auto-immunity. γδ T cells are recognized as IL-17 producers, but based on the level of CD3 expression, we now define the remarkable ability of a CD3(bright) γδ T-cell subset with an effector memory phenotype to rapidly produce IL-17A, but not interferon-γ. CD3(bright) γδ T cells uniformly express the canonical germline encoded Vγ6/Vδ1(+) T-cell receptor. They are widely distributed with a preferential representation in the lungs and skin are negatively impacted in the absence of retinoic acid receptor-related orphan receptor gammat expression or endogenous flora. This population responded rapidly to various stimuli in a mechanism involving IL-23 and NOD-like receptor family, pyrin domain containing 3 (NLRP3)-inflammasome-dependent IL-1ß. Finally, we demonstrated that IL-17-producing CD3(bright) γδ T cells responded promptly and strongly to pneumococcal infection and during skin inflammation. Here, we propose a new way to specifically analyze IL-17-producing Vγ6/Vδ1(+) T cells based on the level of CD3 signals. Using this gating strategy, our data reinforce the crucial role of this γδ T-cell subset in respiratory and skin disorders.


Subject(s)
CD3 Complex/metabolism , Interleukin-17/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Amino Acid Sequence , Aminoquinolines/pharmacology , Animals , CD3 Complex/chemistry , Carrier Proteins/metabolism , Germ Cells/drug effects , Homeostasis/drug effects , Imiquimod , Immunity , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Interleukin-23 , Lung/drug effects , Lung/immunology , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Male , Mice, Inbred C57BL , Molecular Sequence Data , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Skin/drug effects , Skin/immunology , T-Lymphocytes/drug effects
2.
Ann Oncol ; 25(8): 1544-50, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24608200

ABSTRACT

BACKGROUND: We have previously shown the prognostic importance of tumor-infiltrating lymphocytes (TILs) in newly diagnosed triple-negative breast cancer (TNBC) using tumor samples from a large clinical trial cohort. In this study, we aimed to validate these findings and also investigate associations with trastuzumab benefit in HER2-overexpressing disease (HER2+). PATIENTS AND METHODS: A prospective-retrospective study was conducted using samples from the FinHER adjuvant, phase III trial that enrolled 1010 early-stage BC patients, 778 of whom were HER2-nonamplified. Those with HER2+ disease (n = 232) were randomized to 9 weeks of trastuzumab or no trastuzumab in addition to chemotherapy. Two pathologists independently quantified stromal TILs in 935 (92.6%) available slides. The primary end point of distant disease-free survival (DDFS) and interactions with trastuzumab were studied in Cox regression models. RESULTS: Confirming our previous findings, in TNBC (n = 134) each 10% increase in TILs was significantly associated with decreased distant recurrence in TNBC; for DDFS the hazard ratio adjusted for clinicopathological factors: 0.77; 95% confidence interval (CI) 0.61-0.98, P = 0.02. In HER2+ BC (n = 209), each 10% increase in lymphocytic infiltration was significantly associated with decreased distant recurrence in patients randomized to the trastuzumab arm (DDFS P interaction = 0.025). CONCLUSIONS: Higher levels of TILs present at diagnosis were significantly associated with decreased distant recurrence rates in primary TNBC. These results confirm our previous data and further support that TILs should be considered as a robust prognostic factor in this BC subtype. We also report for the first time an association between higher levels of TILs and increased trastuzumab benefit in HER2+ disease. Further research into why some TN and HER2+ BCs can or cannot generate a host antitumor immune response and how trastuzumab can favorably alter the immune microenvironment is warranted.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Pharmacological , Biomarkers, Tumor , Lymphocytes, Tumor-Infiltrating/pathology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology , Adult , Aged , Biomarkers, Pharmacological/analysis , Biomarkers, Tumor/analysis , Chemotherapy, Adjuvant , Female , Finland , Humans , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Prognosis , Trastuzumab , Treatment Outcome , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/pathology
3.
Nat Med ; 7(1): 94-100, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135622

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in various tumor cells in vitro, but its physiological role in tumor surveillance remains unknown. Here, we report that TRAIL is constitutively expressed on murine natural killer (NK) cells in the liver and plays a substantial role in suppressing tumor metastasis. Freshly isolated NK cells, but not natural killer T cells or ordinary T cells, from the liver expressed cell surface TRAIL, which was responsible for spontaneous cytotoxicity against TRAIL-sensitive tumor cells in vitro along with perforin and Fas ligand (FasL). Administration of neutralizing monoclonal antibody against TRAIL significantly increased experimental liver metastases of several TRAIL-sensitive tumor cell lines. Such an anti-metastatic effect of TRAIL was not observed in NK cell-depleted mice or interferon-gamma-deficient mice, the latter of which lacked TRAIL on liver NK cells. These findings provide the first evidence for the physiological function of TRAIL as a tumor suppressor.


Subject(s)
Apoptosis/physiology , Killer Cells, Natural/immunology , Liver/immunology , Membrane Glycoproteins/physiology , Neoplasm Metastasis/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins , Ligands , Liver/cytology , Liver Neoplasms, Experimental/secondary , Male , Mice , Mice, Inbred Strains , TNF-Related Apoptosis-Inducing Ligand
4.
Gene Ther ; 17(9): 1105-16, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20428216

ABSTRACT

The anti-tumor efficacy of adoptively transferred T cells requires their in vivo persistence and memory polarization. It is unknown if human chimeric antigen receptor (CAR)-expressing T cells can also undergo memory polarization. We examined the functional status of CAR CD8(+) T cells, re-directed to Lewis Y antigen (LeY-T), throughout a period of ex vivo expansion. Immediately before culture CD8(+) T cells comprised a mixture of phenotypes including naive (CD45RA(+)/CCR7(+)/CD27(+)/CD28(+)/perforin-), central memory (CM, CD45RA(-)/CCR7(lo)/CD27(+)/CD28(+)/perforin(lo)), effector memory (EM, CD45RA(-)/CCR7(-)/CD27(+)/CD28(+)/perforin(mod)) and effector (Eff, CD45RA(+)/CCR7(-)/CD27(-)/CD28(-)/perforin(hi)) cells. After transduction and expansion culture of peripheral blood mononuclear cells from normal donors or multiple myeloma patients, CD8(+) LeY-T cells polarized to EM- and CM-like phenotype. CD8(+) LeY-T cells differed from starting CD8(+) CM and EM T cells in that CD27, but not CD28, was downregulated. In addition, CD8(+) LeY-T cells expressed high levels of perforin, similar to starting CD8(+) Eff. CD8(+) LeY-T cells also showed hallmarks of both memory and Eff function, underwent homeostatic proliferation in response to interleukin (IL)-15, and showed interferon (IFN)-γ production and cytotoxicity in response to Le-Y antigen on OVCAR-3 (human ovarian adenocarcinoma) cells. This study confirms CD8(+) LeY-T cells have a CM- and EM-like phenotype and heterogeneous function consistent with potential to persist in vivo after adoptive transfer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Immunologic Memory , Receptors, Antigen/genetics , CD28 Antigens/immunology , Cell Proliferation , Humans , Interferon-gamma/metabolism , Leukocyte Common Antigens/immunology , Phenotype , Receptors, Antigen/immunology , Receptors, Antigen/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
5.
Gene Ther ; 17(5): 678-86, 2010 May.
Article in English | MEDLINE | ID: mdl-20200563

ABSTRACT

We have evaluated the carbohydrate antigen Lewis(Y) (Le(Y)) as a potential target for T-cell immunotherapy of hematological neoplasias. Analysis of 81 primary bone marrow samples revealed moderate Le(Y) expression on plasma cells of myeloma patients and myeloblasts of patients with acute myeloid leukemia (AML) (52 and 46% of cases, respectively). We developed a retroviral vector construct encoding a chimeric T-cell receptor that recognizes the Le(Y) antigen in a major histocompatibility complex-independent manner and delivers co-stimulatory signals to achieve T-cell activation. We have shown efficient transduction of peripheral blood-derived T cells with this construct, resulting in antigen-restricted interferon-gamma secretion and cell lysis of Le(Y)-expressing tumor cells. In vivo activity of gene-modified T cells was demonstrated in the delayed growth of myeloma xenografts in NOD/SCID mice, which prolonged survival. Therefore, targeting Le(Y)-positive malignant cells with T cells expressing a chimeric receptor recognizing Le(Y) was effective both in vitro and in a myeloma mouse model. Consequently, we plan to use T cells manufactured under Good Manufacturing Practice conditions in a phase I immunotherapy study for patients with Le(Y)-positive myeloma or AML.


Subject(s)
Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/therapy , Lewis Blood Group Antigens/immunology , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/transplantation , Animals , Female , Genetic Vectors , Humans , Interferon-gamma/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen, T-Cell/immunology , Retroviridae/genetics , T-Lymphocytes/immunology , Transduction, Genetic
6.
J Exp Med ; 190(9): 1297-308, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10544201

ABSTRACT

Optimal T cell differentiation into effector cells with specialized functions requires the participation of cytokine receptor signals. In T helper cells, this process is controlled by chromatin changes and distal and proximal regulatory elements as well as specific transcription factors. Analogous events during cytotoxic T lymphocyte (CTL) differentiation remain to be identified. This process is known, however, to be crucially regulated by interleukin (IL)-2 receptor (R) signals. It is accompanied by the induction of perforin expression via a mechanism that does not entail proximal regulatory elements. In this report, transgenically expressed human perforin gene locus DNAs demonstrate that IL-2R signals target two IL-2-dependent enhancers approximately 15 and 1 kilobase upstream of the promoter. The most distal enhancer may also respond to TCR signals. In transient transfections, both enhancers required two identically spaced Stat-like elements for their activation, which was abolished by expression of a dominant negative signal transducer and activator of transcription (Stat)5 molecule, whereas a constitutively active Stat5 molecule bypassed the requirement for IL-2R signals. These results provide a molecular explanation for the activation of the perforin gene during CTL differentiation and complement the analysis of animals deficient in the activation of the IL-2R Stat signaling pathway by establishing perforin as a target gene.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Membrane Glycoproteins/genetics , Milk Proteins , Receptors, Interleukin-2/metabolism , Signal Transduction/genetics , Trans-Activators/metabolism , Animals , Base Sequence , Cell Line , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Interleukin-2/pharmacology , Mice , Mice, Transgenic , Molecular Sequence Data , Perforin , Pore Forming Cytotoxic Proteins , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid , STAT5 Transcription Factor , T-Lymphocytes/metabolism , Transcriptional Activation , Transfection
7.
J Exp Med ; 188(9): 1611-9, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9802973

ABSTRACT

Natural killer (NK) cells are thought to provide the first line of defence against tumors, particularly major histocompatibility complex (MHC) class I- variants. We have confirmed in C57BL/6 (B6) mice lacking perforin that peritoneal growth of MHC class I- RMA-S tumor cells in unprimed mice is controlled by perforin-dependent cytotoxicity mediated by CD3(-) NK1.1(+) cells. Furthermore, we demonstrate that B6 mice lacking tumor necrosis factor (TNF) are also significantly defective in their rejection of RMA-S, despite the fact that RMA-S is insensitive to TNF in vitro and that spleen NK cells from B6 and TNF-deficient mice are equally lytic towards RMA-S. NK cell recruitment into the peritoneum was abrogated in TNF-deficient mice challenged with RMA-S or RM-1, a B6 MHC class I- prostate carcinoma, compared with B6 or perforin-deficient mice. The reduced NK cell migration to the peritoneum of TNF-deficient mice correlated with the defective NK cell response to tumor in these mice. By contrast, a lack of TNF did not affect peptide-specific cytotoxic T lymphocyte-mediated rejection of tumor from the peritoneum of preimmunized mice. Overall, these data show that NK cells delivering perforin are the major effectors of class I- tumor rejection in the peritoneum, and that TNF is specifically critical for their recruitment to the peritoneum.


Subject(s)
Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Peritoneum/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cell Movement/immunology , Cytotoxicity, Immunologic , Graft Rejection/immunology , Histocompatibility Antigens Class I/metabolism , In Vitro Techniques , Killer Cells, Natural/physiology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neoplasm Transplantation , Perforin , Pore Forming Cytotoxic Proteins , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
8.
J Exp Med ; 172(6): 1877-80, 1990 Dec 01.
Article in English | MEDLINE | ID: mdl-2124255

ABSTRACT

The cytotoxic activity and pore-forming protein (PFP) expression of human peripheral blood (PB) gamma/delta T cells were examined. Fresh gamma/delta T cells isolated from PB lymphocytes by fluorescence-activated cell sorting exhibited a substantial natural killer-like cytotoxic activity against K562 target cells and had a high cytotoxic potential triggered by anti-CD3 monoclonal antibody (mAb) against P815 target cells bearing Fc gamma R. Immunocytochemical staining with an anti-PFP mAb revealed that virtually all PB gamma/delta T cells are granular lymphocytes with abundant PFP in their cytoplasmic granules. Constitutive expression of PFP in PB gamma/delta T cells was also demonstrated by Northern blot analysis. These observations support the proposed role of gamma/delta T cells in cytolytic immune surveillance in vivo.


Subject(s)
Cytotoxicity, Immunologic , Membrane Glycoproteins , Membrane Proteins/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Antibodies, Monoclonal , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD8 Antigens , Cells, Cultured , Humans , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Membrane Proteins/analysis , Perforin , Pore Forming Cytotoxic Proteins
9.
J Exp Med ; 171(4): 1269-81, 1990 Apr 01.
Article in English | MEDLINE | ID: mdl-1691263

ABSTRACT

Our studies have analyzed pore-forming protein (PFP) mRNA expression in resting and stimulated human peripheral blood CD3- large granular lymphocytes (LGL), CD3+ T cells, and their CD4+ or CD8+ subsets. Signals that stimulate T cells to develop cytotoxic activity (i.e., IL-2 or OKT-3 mAb) led to the induction of PFP mRNA in T cells. The data indicated that IL-2 directly increased PFP mRNA in the CD8+ subset of T cells, in the absence of new DNA or protein synthesis. Abrogation of IL-2-induced PFP mRNA expression and cytotoxic potential of T cells by the anti-p75 IL-2 receptor mAb suggested that low numbers of p75 IL-2 receptors on CD8+ T cells were capable of transducing signals responsible for these IL-2-induced effects. The induction of T cell PFP mRNA via CD3, using OKT-3 mAb, was less rapid but greater than that caused by IL-2; however, a combination of PMA and ionomycin, which bypasses crosslinking of the TCR/CD3 complex, could not mimic this increase in PFP mRNA levels in T cells. The role of second messenger systems in regulating PFP mRNA expression remains to be determined. In contrast, high constitutive PFP mRNA expression was observed in CD3- LGL and these mRNA levels could not be enhanced by stimulation with IL-2. The cytotoxic potential of peripheral blood T cells and LGL induced in response to IL-2 correlated with IL-2-induced PFP mRNA levels in these cells and was consistent with PFP being one of several important molecules involved in the effector function of cytotoxic lymphocytes.


Subject(s)
Gene Expression/drug effects , Interleukin-2/pharmacology , Membrane Glycoproteins , Membrane Proteins/genetics , T-Lymphocytes, Cytotoxic/metabolism , Antibodies, Monoclonal , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD3 Complex , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Interferon-gamma/biosynthesis , Kinetics , Lymphocyte Activation , Perforin , Pore Forming Cytotoxic Proteins , RNA/blood , RNA/genetics , RNA/isolation & purification , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
10.
J Exp Med ; 192(5): 755-60, 2000 Sep 04.
Article in English | MEDLINE | ID: mdl-10974040

ABSTRACT

Immune surveillance by cytotoxic lymphocytes against cancer has been postulated for decades, but direct evidence for the role of cytotoxic lymphocytes in protecting against spontaneous malignancy has been lacking. As the rejection of many experimental cancers by cytotoxic T lymphocytes and natural killer cells is dependent on the pore-forming protein perforin (pfp), we examined pfp-deficient mice for increased cancer susceptibility. Here we show that pfp-deficient mice have a high incidence of malignancy in distinct lymphoid cell lineages (T, B, NKT), indicating a specific requirement for pfp in protection against lymphomagenesis. The susceptibility to lymphoma was accentuated by simultaneous lack of expression of the p53 gene, mutations in which also commonly predispose to human malignancies, including lymphoma. In contrast, the incidence and age of onset of sarcoma was unaffected in p53-deficient mice. Pfp-deficient mice were at least 1,000-fold more susceptible to these lymphomas when transplanted, compared with immunocompetent mice in which tumor rejection was controlled by CD8(+) T lymphocytes. This study is the first that implicates direct cytotoxicity by lymphocytes in regulating lymphomagenesis.


Subject(s)
Cytotoxicity, Immunologic , Lymphoma/etiology , Membrane Glycoproteins/physiology , Animals , Graft Rejection , Humans , Lymphoma/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Perforin , Pore Forming Cytotoxic Proteins , Tumor Suppressor Protein p53/physiology
11.
J Exp Med ; 193(6): 661-70, 2001 Mar 19.
Article in English | MEDLINE | ID: mdl-11257133

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is expressed by in vitro activated natural killer (NK) cells, but the relevance of this observation to the biological function of NK cells has been unclear. Herein, we have demonstrated the in vivo induction of mouse TRAIL expression on various tissue NK cells and correlated NK cell activation with TRAIL-mediated antimetastatic function in vivo. Expression of TRAIL was only constitutive on a subset of liver NK cells, and innate NK cell control of Renca carcinoma hepatic metastases in the liver was partially TRAIL dependent. Administration of therapeutic doses of interleukin (IL)-12, a powerful inducer of interferon (IFN)-gamma production by NK cells and NKT cells, upregulated TRAIL expression on liver, spleen, and lung NK cells, and IL-12 suppressed metastases in both liver and lung in a TRAIL-dependent fashion. By contrast, alpha-galactosylceramide (alpha-GalCer), a powerful inducer of NKT cell IFN-gamma and IL-4 secretion, suppressed both liver and lung metastases but only stimulated NK cell TRAIL-mediated function in the liver. TRAIL expression was not detected on NK cells from IFN-gamma-deficient mice and TRAIL-mediated antimetastatic effects of IL-12 and alpha-GalCer were strictly IFN-gamma dependent. These results indicated that TRAIL induction on NK cells plays a critical role in IFN-gamma-mediated antimetastatic effects of IL-12 and alpha-GalCer.


Subject(s)
Carcinoma, Renal Cell/secondary , Interferon-gamma/immunology , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Liver Neoplasms/secondary , Membrane Glycoproteins/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Apoptosis Regulatory Proteins , Carcinoma, Renal Cell/immunology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Galactosylceramides/immunology , Galactosylceramides/pharmacology , Humans , Interleukin-12/immunology , Interleukin-12/pharmacology , Interleukin-2/immunology , Interleukin-2/pharmacology , Kidney Neoplasms/pathology , Kidney Neoplasms/prevention & control , Ligands , Liver Neoplasms/immunology , Liver Neoplasms/prevention & control , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , TNF-Related Apoptosis-Inducing Ligand , Tissue Distribution , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis
12.
J Exp Med ; 190(3): 435-44, 1999 Aug 02.
Article in English | MEDLINE | ID: mdl-10430631

ABSTRACT

Until now, it has been unclear whether murine cytomegalovirus (MCMV)-encoded protein m144 directly regulates natural killer (NK) cell effector function and whether the effects of m144 are only strictly evident in the context of MCMV infection. We have generated clones of the transporter associated with antigen processing (TAP)-2-deficient RMA-S T lymphoma cell line and its parent cell line, RMA, that stably express significant and equivalent levels of m144. In vivo NK cell-mediated rejection of RMA-S-m144 lymphomas was reduced compared with rejection of parental or mock-transfected RMA-S clones, indicating the ability of m144 to regulate NK cell-mediated responses in vivo. Significantly, the accumulation of NK cells in the peritoneum was reduced in mice challenged with RMA-S-m144, as was the lytic activity of NK cells recovered from the peritoneum. Expression of m144 on RMA-S cells also conferred resistance to cytotoxicity mediated in vitro by interleukin 2-activated adherent spleen NK cells. In summary, the data demonstrate that m144 confers some protection from NK cell effector function mediated in the absence of target cell class I expression, but that in vivo the major effect of m144 is to regulate NK cell accumulation and activation at the site of immune challenge.


Subject(s)
Graft Rejection/immunology , Histocompatibility Antigens Class I , Killer Cells, Natural/immunology , Lymphoma, T-Cell/immunology , Muromegalovirus/immunology , Viral Proteins/physiology , Animals , Cell Movement/immunology , Cytotoxicity, Immunologic/genetics , Graft Rejection/genetics , Histocompatibility Antigens Class I/genetics , Immunity, Innate , Killer Cells, Natural/virology , Lymphocyte Activation , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Sequence Homology, Amino Acid , Transfection , Tumor Cells, Cultured , Viral Proteins/biosynthesis , Viral Proteins/immunology
13.
J Exp Med ; 191(1): 89-96, 2000 Jan 03.
Article in English | MEDLINE | ID: mdl-10620607

ABSTRACT

Tumor necrosis factor (TNF) and Fas ligand (FasL) play major roles in the homeostasis of the peripheral immune system. This becomes dramatically obvious in the absence of a functional FasL. Mice with such a deficiency develop a profound lymphadenopathy, splenomegaly, hypergammaglobulinemia, and strain-dependent systemic autoimmune disease, and succumb to premature death. It is consequently termed generalized lymphoproliferative disorder (gld). By contrast, TNF deficiency alone does not result in a striking phenotype. Thus, we sought to determine what role TNF might play in contributing to the gld phenotype by creating C57BL/6.gld.TNF(-/-) mice. Contrary to the expected outcome, mice deficient for both FasL and TNF had a substantially milder gld phenotype with regard to mortality, lymphoaccumulation, germinal center formation, and hypergammaglobulinemia. To confirm these data in a strain highly permissive for the phenotype, C3H/HeJ.gld and C3H.HeJ.lpr mice were treated with a TNF-specific monoclonal antibody. This transient neutralization of TNF also resulted in a significantly attenuated lymphoproliferative phenotype. We conclude that TNF is necessary for the full manifestation of the lymphoproliferative disorder, in particular playing a critical role in lymphoaccumulation. Most importantly, absence of TNF protects gld mice against premature death.


Subject(s)
Lymphoproliferative Disorders/etiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/metabolism , Fas Ligand Protein , Germinal Center/physiology , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C3H , Mice, Knockout , Phenotype , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type II
14.
J Exp Med ; 191(4): 661-8, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10684858

ABSTRACT

Natural tumor surveillance capabilities of the host were investigated in six different mouse tumor models where endogenous interleukin (IL)-12 does or does not dictate the efficiency of the innate immune response. Gene-targeted and lymphocyte subset-depleted mice were used to establish the relative importance of natural killer (NK) and NK1.1(+) T (NKT) cells in protection from tumor initiation and metastasis. In the models examined, CD3(-) NK cells were responsible for tumor rejection and protection from metastasis in models where control of major histocompatibility complex class I-deficient tumors was independent of IL-12. A protective role for NKT cells was only observed when tumor rejection required endogenous IL-12 activity. In particular, T cell receptor Jalpha281 gene-targeted mice confirmed a critical function for NKT cells in protection from spontaneous tumors initiated by the chemical carcinogen, methylcholanthrene. This is the first description of an antitumor function for NKT cells in the absence of exogenously administered potent stimulators such as IL-12 or alpha-galactosylceramide.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-12/physiology , Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Receptors, Antigen, T-Cell, alpha-beta/physiology , T-Lymphocyte Subsets/immunology , Animals , Crosses, Genetic , Female , Galactosylceramides/pharmacology , Genes, T-Cell Receptor alpha , Interleukin-12/pharmacology , Liver/immunology , Male , Methylcholanthrene , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/prevention & control , Receptor-CD3 Complex, Antigen, T-Cell/deficiency , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymus Gland/immunology , Tumor Cells, Cultured
15.
Trends Biochem Sci ; 25(1): 1-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10637601

ABSTRACT

Multidrug resistance mediated by the drug-efflux protein P-glycoprotein (P-gp) is one mechanism that tumor cells use to escape death induced by chemotherapeutic drugs. Although it is irrefutable that P-gp can efflux xenobiotics out of cells, biological regulatory functions for P-gp in multicellular organisms have yet to be established firmly. Recent observations have challenged the notion that P-gp has evolved merely to efflux xenotoxins out of healthy cells and raised the possibility that P-gp and related transporter molecules might play a fundamental role in regulating cell differentiation, proliferation and survival.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Cell Death/physiology , Cell Differentiation/physiology , Animals , Biological Transport , Cholesterol/metabolism , Drug Resistance, Multiple/physiology , Humans , Lipid Metabolism
16.
Oncogene ; 26(25): 3745-57, 2007 May 28.
Article in English | MEDLINE | ID: mdl-17530027

ABSTRACT

Deregulated cell death pathways may lead to the development of cancer, and induction of tumor cell apoptosis is the basis of many cancer therapies. Knowledge accumulated concerning the molecular mechanisms of apoptotic cell death has aided the development of new therapeutic strategies to treat cancer. Signals through death receptors of the tumor necrosis factor (TNF) superfamily have been well elucidated, and death receptors are now one of the most attractive therapeutic targets in cancer. In particular, DR5 and DR4, death receptors of TNF-related apoptosis-inducing ligand (TRAIL or Apo2L), are interesting targets of antibody-based therapy, since TRAIL may also bind decoy receptors that may prevent TRAIL-mediated apoptosis, whereas TRAIL ligand itself selectively induces apoptosis in cancer cells. Here, we review the potential therapeutic utility of agonistic antibodies against DR5 and DR4 and discuss the possible extension of this single-antibody-based strategy when combined with additional modalities that either synergizes to cause enhanced apoptosis or further engage the cellular immune response. Rational design of antibody-based therapies combining the induction of tumor cell apoptosis and activation of tumor-specific adaptive immunity enables promotion of distinct steps of the antitumor immune response, thereby enhancing tumor-specific lymphocytes that can eradicate TRAIL/DR5-resistant mutating, large established and heterogeneous tumors in a manner that does not require the definition of individual tumor-specific antigens.


Subject(s)
Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, Death Domain/metabolism , Animals , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Disease Progression , Humans , Immunotherapy , Neoplasms/immunology , Neoplasms/pathology , Receptors, Death Domain/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism
17.
Gene Ther ; 15(14): 1056-66, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18369322

ABSTRACT

There is considerable concern regarding the transforming potential of retroviral vectors currently used for gene therapy, with evidence that retroviral integration can lead to leukemia in recipients of gene-modified stem cells. However, it is not clear whether retroviral-mediated transduction of T cells can lead to malignancy. We transduced mouse T cells with a Moloney murine retroviral gene construct and transferred them into congenic mice, which were preconditioned to enhance the engraftment of transferred T cells. Recipients were then observed long-term for evidence of cancer. Transferred T cells persisted in mice throughout life at levels up to 17% with gene copy numbers up to 5.89 x 10(5) per million splenocytes. Mice receiving gene-modified T cells developed tumors at a similar rate as control mice that did not receive T cells, and tumors in both groups of mice were of a similar range of histologies. Hematological malignancies comprised approximately 60% of cancers, and the remaining cancers consisted largely of carcinomas. Importantly, the incidence of lymphomas was similar in both groups of mice, and no lymphomas were found to be of donor T-cell origin. This study indicates that the use of retroviral vectors to transduce T cells does not lead to malignant transformation.


Subject(s)
Adoptive Transfer , Genetic Therapy/adverse effects , Genetic Vectors/administration & dosage , Moloney murine leukemia virus/physiology , T-Lymphocytes/virology , Animals , Cell Transformation, Viral , Leukemia/virology , Lymphoma/virology , Mice , Mice, SCID , Moloney murine leukemia virus/genetics , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/transplantation , Time , Transduction, Genetic/methods , Transgenes
18.
Curr Top Microbiol Immunol ; 314: 293-323, 2007.
Article in English | MEDLINE | ID: mdl-17593666

ABSTRACT

CD1d-restricted T cells (NKT cells) are potent regulators of a broad range of immune responses. In particular, an abundance of research has focussed on the role of NKT cells in tumor immunity. This field of research has been greatly facilitated by the finding of agonist ligands capable of potently stimulating NKT cells and also animal models where NKT cells have been shown to play a natural role in the surveillance of tumors. Herein, we review the capability of NKT cells to promote the rejection of tumors and the mechanisms by which this occurs. We also highlight a growing field of research that has found that NKT cells are capable of suppressing anti-tumor immunity and discuss the progress to date for the immunotherapeutic use of NKT cells.


Subject(s)
Antigens, CD1/metabolism , Killer Cells, Natural/immunology , Neoplasms/immunology , Animals , Clinical Trials, Phase I as Topic , Disease Models, Animal , Galactosylceramides/immunology , Galactosylceramides/therapeutic use , Humans , Killer Cells, Natural/metabolism , Mice , Monitoring, Immunologic , Neoplasms/therapy
19.
J Clin Invest ; 102(2): 329-39, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9664074

ABSTRACT

Both p53 and ceramide have been implicated in the regulation of growth suppression. p53 has been proposed as the "guardian of the genome" and ceramide has been suggested as a "tumor suppressor lipid. " Both molecules appear to regulate cell cycle arrest, senescence, and apoptosis. In this study, we investigated the relationship between p53 and ceramide. We found that treatment of Molt-4 cells with low concentrations of actinomycin D or gamma-irradiation, which activate p53-dependent apoptosis, induces apoptosis only in cells expressing normal levels of p53. In these cells, p53 activation was followed by a dose- and time-dependent increase in endogenous ceramide levels which was not seen in cells lacking functional p53 and treated similarly. Similar results were seen in irradiated L929 cells whereby the p53-deficient clone was significantly more resistant to irradiation and exhibited no ceramide response. However, in p53-independent systems, such as growth suppression induced by TNF-alpha or serum deprivation, ceramide accumulated irrespective of the upregulation of p53, indicating that p53 regulates ceramide accumulation in only a subset of growth-suppressive pathways. Finally, ceramide did not increase p53 levels when used at growth-suppressive concentrations. Also, when cells lacking functional p53, either due to mutation or the expression of the E6 protein of human papilloma virus, were treated with exogenous ceramide, there was equal growth suppression, cell cycle arrest, and apoptosis as compared with cells expressing normal p53. These results indicate that p53 is unlikely to function "downstream" of ceramide. Instead, they suggest that, in situations where p53 performs a critical regulatory role, such as the response to genotoxic stress, it functions "upstream" of ceramide. These studies begin to define a relationship between these two pathways of growth inhibition.


Subject(s)
Apoptosis , Ceramides/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Dactinomycin/pharmacology , Evaluation Studies as Topic , Gamma Rays , HL-60 Cells , Humans , Mice , Mutagenicity Tests , Tumor Cells, Cultured
20.
Cancer Gene Ther ; 14(5): 499-508, 2007 May.
Article in English | MEDLINE | ID: mdl-17332777

ABSTRACT

Activation and expansion of T cells are important in disease resolution, but tumors do not usually satisfy these immune requirements. Therefore, we employed a novel strategy whereby dual-specific T cells were generated that could respond to both tumor and influenza virus, reasoning that immunization with influenza virus would activate and expand tumor-specific cells, and inhibit tumor growth. Dual-specific T cells were generated by gene modification of influenza virus-specific mouse T cells with a chimeric gene-encoding reactivity against the erbB2 tumor-associated antigen. Dual-specific T cells were demonstrated to respond against both tumor and influenza in vitro, and expanded in vitro in response to influenza to a much greater degree than in response to tumor cells. Following adoptive transfer and immunization of tumor-bearing mice with influenza virus, dual-specific T cells expanded greatly in numbers in the peritoneal cavity and spleen. This resulted in a significant increase in time of survival of mice. However, tumors were not eradicated, which may have been due to the observed poor penetration of tumor by T cells. This is the first demonstration that the potent immunogenic nature of an infectious agent can be utilized to directly impact on T-cell expansion and activity against tumor in vivo.


Subject(s)
Immunotherapy, Adoptive/methods , Mammary Neoplasms, Animal/therapy , Orthomyxoviridae/immunology , Receptor, ErbB-2/antagonists & inhibitors , T-Lymphocytes/transplantation , Animals , Antigens, Neoplasm/immunology , Mice , Mice, Inbred Strains , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL