Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
2.
ESMO Open ; 7(2): 100455, 2022 04.
Article in English | MEDLINE | ID: mdl-35398718

ABSTRACT

BACKGROUND: Chemoradiotherapy with durvalumab consolidation has yielded excellent results in stage III non-small-cell lung cancer (NSCLC). Therefore, it is essential to identify patients who might benefit from a surgical approach. MATERIAL AND METHODS: Data from 437 patients with operable stage III NSCLC enrolled in four consecutive Swiss Group for Clinical Cancer Research (SAKK) trials (16/96, 16/00, 16/01, 16/08) were pooled and outcomes were analyzed in 431 eligible patients. All patients were treated with three cycles of induction chemotherapy (cisplatin/docetaxel), followed in some patients by neoadjuvant radiotherapy (44 Gy, 22 fractions) (16/00, 16/01, 16/08) and cetuximab (16/08). RESULTS: With a median follow-up time of 9.3 years (range 8.5-10.3 years), 5- and 10-year overall survival (OS) rates were 37% and 25%, respectively. Overall, 342 patients (79%) underwent tumor resection, with a complete resection (R0) rate of 80%. Patients (n = 272, 63%) with R0 had significantly longer OS compared to patients who had surgery but incomplete resection (64.8 versus 19.2 months, P < 0.001). OS for patients who achieved pathological complete remission (pCR) (n = 66, 15%) was significantly better compared to resected patients without pCR (86.5 versus 37.0 months, P = 0.003). For patients with pCR, the 5- and 10-year event-free survival and OS rates were 45.7% [95% confidence interval (CI) 32.8% to 57.7%] and 28.1% (95% CI 15.2% to 42.6%), and 58.2% (95% CI 45.2% to 69.2%) and 45.0% (95% CI 31.5% to 57.6%), respectively. CONCLUSION: We report favorable long-term outcomes in patients with operable stage III NSCLC treated with neoadjuvant chemotherapy with cisplatin and docetaxel ± neoadjuvant sequential radiotherapy from four prospective SAKK trials. Almost two-third of the patients underwent complete resection after neoadjuvant therapy. We confirm R0 resection and pCR as important predictors of outcome.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/therapeutic use , Docetaxel/pharmacology , Docetaxel/therapeutic use , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Neoplasm Staging , Prospective Studies
3.
Nat Commun ; 12(1): 1065, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594067

ABSTRACT

The production of blood cells during steady-state and increased demand depends on the regulation of hematopoietic stem cell (HSC) self-renewal and differentiation. Similarly, the balance between self-renewal and differentiation of leukemia stem cells (LSCs) is crucial in the pathogenesis of leukemia. Here, we document that the TNF receptor superfamily member lymphotoxin-ß receptor (LTßR) and its ligand LIGHT regulate quiescence and self-renewal of murine and human HSCs and LSCs. Cell-autonomous LIGHT/LTßR signaling on HSCs reduces cell cycling, promotes symmetric cell division and prevents primitive HSCs from exhaustion in serial re-transplantation experiments and genotoxic stress. LTßR deficiency reduces the numbers of LSCs and prolongs survival in a murine chronic myeloid leukemia (CML) model. Similarly, LIGHT/LTßR signaling in human G-CSF mobilized HSCs and human LSCs results in increased colony forming capacity in vitro. Thus, our results define LIGHT/LTßR signaling as an important pathway in the regulation of the self-renewal of HSCs and LSCs.


Subject(s)
Cell Differentiation , Cell Self Renewal , Hematopoietic Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Lymphotoxin beta Receptor/metabolism , Neoplastic Stem Cells/pathology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Antigens, CD34/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , DNA Damage , Fluorouracil/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects
4.
Cancer Immunol Immunother ; 70(2): 405-415, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32767058

ABSTRACT

BACKGROUND: Over the past few years, immune checkpoint inhibitors have changed the therapeutic landscape of non-small-cell lung cancer (NSCLC). Response to immune checkpoint inhibitors correlates with a pre-existing anti-tumoral immune response. Checkpoint inhibitors have been introduced as second-line therapy and are only very recently used as monotherapy or in combination with chemotherapy as first-line treatment of NSCLC. However, the effect of conventional first-line platinum-based chemotherapy on the immune infiltrate in the tumor is largely unknown. METHODS: We measured the gene expression of a custom set of 201 cancer- and immune-related genes in 100 NSCLC tumor biopsies collected before chemotherapy and 33 re-biopsies after platinum-based chemotherapy at the time point of progression. For 29 patients matched pre- and post-chemotherapy samples could be evaluated. RESULTS: We identified a cluster of 47 co-expressed immune genes, including PDCD1 (PD1) and CD274 (PD-L1), along with three other co-expression clusters. Chemotherapy decreased the average gene expression of the immune cluster while no effect was observed on the other three cluster. Within this immune cluster, CTLA4, LAG3, TNFRSF18, CD80 and FOXP3 were found to be significantly decreased in patient-matched samples after chemotherapy. CONCLUSION: Our results suggest that conventional platinum-based chemotherapy negatively impacts the immune microenvironment at the time point of secondary progression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/surgery , Gene Expression/genetics , Lung Neoplasms/genetics , Lung Neoplasms/surgery , Tumor Microenvironment/genetics , Biopsy , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Male
5.
Ann Oncol ; 29(6): 1423-1430, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29648580

ABSTRACT

Background: The addition of bevacizumab to temozolomide-based chemoradiotherapy (TMZ/RT â†’ TMZ) did not prolong overall survival (OS) in patients with newly diagnosed glioblastoma in phase III trials. Elderly and frail patients are underrepresented in clinical trials, but early reports suggested preferential benefit in this population. Patients and methods: ARTE was a 2 : 1 randomized, multi-center, open-label, non-comparative phase II trial of hypofractionated RT (40 Gy in 15 fractions) with bevacizumab (10 mg/kg×14 days) (arm A, N = 50) or without bevacizumab (arm B, N = 25) in patients with newly diagnosed glioblastoma aged ≥65 years. The primary objective was to obtain evidence for prolongation of median OS by the addition of bevacizumab to RT. Response was assessed by RANO criteria. Quality of life (QoL) was monitored by the EORTC QLQ-C30/BN20 modules. Exploratory studies included molecular subtyping by 450k whole methylome and gene expression analyses. Results: Median PFS was longer in arm A than in arm B (7.6 and 4.8 months, P = 0.003), but OS was similar (12.1 and 12.2 months, P = 0.77). Clinical deterioration was delayed and more patients came off steroids in arm A. Prolonged PFS in arm A was confined to tumors with the receptor tyrosine kinase (RTK) I methylation subtype (HR 0.25, P = 0.014) and proneural gene expression (HR 0.29, P = 0.025). In a Cox model of OS controlling for established prognostic factors, associations with more favorable outcome were identified for age <70 years (HR 0.52, P = 0.018) and Karnofsky performance score 90%-100% (HR 0.51, P = 0.026). Including molecular subtypes into that model identified an association of the RTK II gene methylation subtype with inferior OS (HR 1.73, P = 0.076). Conclusion: Efficacy outcomes and exploratory analyses of ARTE do not support the hypothesis that the addition of bevacizumab to RT generally prolongs survival in elderly glioblastoma patients. Molecular biomarkers may identify patients with preferential benefit from bevacizumab. Clinical trial registration number: NCT01443676.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Bevacizumab/therapeutic use , Chemoradiotherapy/mortality , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Quality of Life , Radiation Dose Hypofractionation , Aged , Aged, 80 and over , Female , Follow-Up Studies , Glioblastoma/pathology , Humans , Male , Prognosis , Survival Rate
7.
Cell Death Differ ; 22(2): 187-98, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24992931

ABSTRACT

Hematopoietic stem cells (HSCs) are rare, multipotent cells that generate via progenitor and precursor cells of all blood lineages. Similar to normal hematopoiesis, leukemia is also hierarchically organized and a subpopulation of leukemic cells, the leukemic stem cells (LSCs), is responsible for disease initiation and maintenance and gives rise to more differentiated malignant cells. Although genetically abnormal, LSCs share many characteristics with normal HSCs, including quiescence, multipotency and self-renewal. Normal HSCs reside in a specialized microenvironment in the bone marrow (BM), the so-called HSC niche that crucially regulates HSC survival and function. Many cell types including osteoblastic, perivascular, endothelial and mesenchymal cells contribute to the HSC niche. In addition, the BM functions as primary and secondary lymphoid organ and hosts various mature immune cell types, including T and B cells, dendritic cells and macrophages that contribute to the HSC niche. Signals derived from the HSC niche are necessary to regulate demand-adapted responses of HSCs and progenitor cells after BM stress or during infection. LSCs occupy similar niches and depend on signals from the BM microenvironment. However, in addition to the cell types that constitute the HSC niche during homeostasis, in leukemia the BM is infiltrated by activated leukemia-specific immune cells. Leukemic cells express different antigens that are able to activate CD4(+) and CD8(+) T cells. It is well documented that activated T cells can contribute to the control of leukemic cells and it was hoped that these cells may be able to target and eliminate the therapy-resistant LSCs. However, the actual interaction of leukemia-specific T cells with LSCs remains ill-defined. Paradoxically, many immune mechanisms that evolved to activate emergency hematopoiesis during infection may actually contribute to the expansion and differentiation of LSCs, promoting leukemia progression. In this review, we summarize mechanisms by which the immune system regulates HSCs and LSCs.


Subject(s)
Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/immunology , Immune System , Leukemia/immunology , Neoplastic Stem Cells/immunology , Stem Cell Niche/immunology , Humans
8.
Lung Cancer ; 85(2): 306-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24928469

ABSTRACT

OBJECTIVES: Molecular subclassification of non small-cell lung cancer (NSCLC) is essential to improve clinical outcome. This study assessed the prognostic and predictive value of circulating micro-RNA (miRNA) in patients with non-squamous NSCLC enrolled in the phase II SAKK (Swiss Group for Clinical Cancer Research) trial 19/05, receiving uniform treatment with first-line bevacizumab and erlotinib followed by platinum-based chemotherapy at progression. MATERIALS AND METHODS: Fifty patients with baseline and 24 h blood samples were included from SAKK 19/05. The primary study endpoint was to identify prognostic (overall survival, OS) miRNA's. Patient samples were analyzed with Agilent human miRNA 8x60K microarrays, each glass slide formatted with eight high-definition 60K arrays. Each array contained 40 probes targeting each of the 1347 miRNA. Data preprocessing included quantile normalization using robust multi-array average (RMA) algorithm. Prognostic and predictive miRNA expression profiles were identified by Spearman's rank correlation test (percentage tumor shrinkage) or log-rank testing (for time-to-event endpoints). RESULTS: Data preprocessing kept 49 patients and 424 miRNA for further analysis. Ten miRNA's were significantly associated with OS, with hsa-miR-29a being the strongest prognostic marker (HR=6.44, 95%-CI 2.39-17.33). Patients with high has-miR-29a expression had a significantly lower survival at 10 months compared to patients with a low expression (54% versus 83%). Six out of the 10 miRNA's (hsa-miRN-29a, hsa-miR-542-5p, hsa-miR-502-3p, hsa-miR-376a, hsa-miR-500a, hsa-miR-424) were insensitive to perturbations according to jackknife cross-validation on their HR for OS. The respective principal component analysis (PCA) defined a meta-miRNA signature including the same 6 miRNA's, resulting in a HR of 0.66 (95%-CI 0.53-0.82). CONCLUSION: Cell-free circulating miRNA-profiling successfully identified a highly prognostic 6-gene signature in patients with advanced non-squamous NSCLC. Circulating miRNA profiling should further be validated in external cohorts for the selection and monitoring of systemic treatment in patients with advanced NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Gene Expression Profiling , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/genetics , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Combined Modality Therapy , Disease Progression , Erlotinib Hydrochloride , Female , Gene Expression , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Male , MicroRNAs/blood , Middle Aged , Neoplasm Staging , Platinum/administration & dosage , Prognosis , Prospective Studies , Quinazolines/administration & dosage , Reproducibility of Results , Risk Factors
9.
Ann Oncol ; 23(2): 531-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21527587

ABSTRACT

BACKGROUND: Oral temozolomide has shown similar efficacy to dacarbazine in phase III trials with median progression-free survival (PFS) of 2.1 months. Bevacizumab has an inhibitory effect on the proliferation of melanoma and sprouting endothelial cells. We evaluated the addition of bevacizumab to temozolomide to improve efficacy in stage IV melanoma. PATIENTS AND METHODS: Previously untreated metastatic melanoma patients with Eastern Cooperative Oncology Group performance status of two or more were treated with temozolomide 150 mg/m(2) days 1-7 orally and bevacizumab 10 mg/kg body weight i.v. day 1 every 2 weeks until disease progression or unacceptable toxicity. The primary end point was disease stabilisation rate [complete response (CR), partial response (PR) or stable disease (SD)] at week 12 (DSR12); secondary end points were best overall response, PFS, overall survival (OS) and adverse events. RESULTS: Sixty-two patients (median age 59 years) enrolled at nine Swiss centres. DSR12 was 52% (PR: 10 patients and SD: 22 patients). Confirmed overall response rate was 16.1% (CR: 1 patient and PR: 9 patients). Median PFS and OS were 4.2 and 9.6 months. OS (12.0 versus 9.2 months; P = 0.014) was higher in BRAF V600E wild-type patients. CONCLUSIONS: The primary end point was surpassed showing promising activity of this bevacizumab/temozolomide combination with a favourable toxicity profile. Response and OS were significantly higher in BRAF wild-type patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Female , Humans , Male , Melanoma/secondary , Middle Aged , Skin Neoplasms/secondary , Temozolomide
10.
Transfusion ; 42(7): 947-53, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12375669

ABSTRACT

BACKGROUND: The quantification of fetal cells in the maternal circulation remains an important goal to determine the amount of anti-D necessary to prevent active immunization of a D- mother giving birth to a D+ baby. Underestimation of fetomaternal hemorrhage (FMH) results in inefficient anti-D prophylaxis and maternal immunization; overestimation of FMH results in higher doses of passively transferred anti-D, higher costs, and the risk of disease transmission. Thus, a reliable method to quantitatively assess FMH is necessary. STUDY DESIGN AND METHODS: Serial dilutions of artificial FMH were quantitatively measured by three different methods: flow cytometry, fluorescence microscopy (each after anti-D staining), and by the Kleihauer-Betke test. The accuracy and precision of the three methods were compared by statistical analysis. RESULTS: Fluorescence microscopy and flow cytometry were comparably accurate and precise in quantifying FMH. In contrast, the accuracy of the Kleihauer-Betke test was poor, resulting in substantial overestimation of FMH in the samples with lower fetal cell concentrations. CONCLUSION: Anti-D flow cytometry and fluorescence microscopy for detection of fetal cells offer equally reliable and precise methods in contrast to the Kleihauer-Betke test. Fluorescence microscopy may be established as standard to quantify FMH in clinical practice because it is comparable to flow cytometry; in addition, it is time saving and is less expensive.


Subject(s)
Fetomaternal Transfusion/diagnosis , Rh-Hr Blood-Group System/blood , Adult , Diagnostic Errors , Female , Fetal Blood , Flow Cytometry/standards , Humans , Infant, Newborn , Microscopy, Fluorescence/standards , Models, Biological , Pregnancy , Reproducibility of Results , Rho(D) Immune Globulin
12.
Transplantation ; 72(4): 712-9, 2001 Aug 27.
Article in English | MEDLINE | ID: mdl-11544436

ABSTRACT

BACKGROUND: Leflunomide is an isoxazol derivative with immunosuppressive capacities in various experimental allo- and xenotransplantation models. Two main mechanisms of action have been described: Inhibition of pyrimidine de novo synthesis and impairment of tyrosine phosphorylation of different tyrosine kinases involved in receptor signaling via B cell and cytokine receptors. MATERIALS AND METHODS: Interference of Leflunomide with the IgM antibody responses to vesicular stomatitis virus (VSV, T-independent type 1), IgM to recombinant VSV glycoprotein (T-independent type 2), and IgG to lymphocytic choriomeningitis virus (LCMV, T-dependent) were analyzed whereas the cytotoxic T cell (CTL) response was examined after LCMV infection. Interference with the CD8+ T cell-mediated autoimmune diabetes in a transgenic mouse expressing the LCMV-glycoprotein in the pancreatic islets was studied as a model for T cell-mediated autoimmune diseases. Uridine substitution experiments were performed to differentiate between the above mentioned two mechanisms of action on different functions of the immune system in vivo. RESULTS: Leflunomide at 35 mg/kg/day suppressed the humoral immune response against all antigens tested. Similar effects on T-independent compared to T-dependent antibody responses required two to four times higher drug doses. CTL responses to LCMV were considerably impaired. Uridine substitution prevented lethal VSV encephalitis under Leflunomide treatment by restoring the neutralizing IgM and IgG responses. However, the inhibition of LCMV specific CTLs and suppression of CD8+ T cell-mediated autoimmune diabetes remained unaffected by additional uridine treatment. CONCLUSIONS: Leflunomide-mediated suppression of B cell and T helper cell activity but not of CTLs largely depends on inhibition of pyrimidine de novo synthesis.


Subject(s)
Antibodies, Viral/analysis , Immunosuppressive Agents/pharmacology , Isoxazoles/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Uridine/pharmacology , Animals , Antibody Formation/drug effects , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus/immunology , Diabetes Mellitus/prevention & control , Dose-Response Relationship, Drug , Leflunomide , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/physiology , Vesicular stomatitis Indiana virus/immunology
13.
Nature ; 411(6841): 1058-64, 2001 Jun 28.
Article in English | MEDLINE | ID: mdl-11429607

ABSTRACT

The vertebrate immune system has evolved to protect against infections that threaten survival before reproduction. Clinically manifest tumours mostly arise after the reproductive years and somatic mutations allow even otherwise antigenic tumours to evade the attention of the immune system. Moreover, the lack of immunological co-stimulatory molecules on solid tumours could result in T-cell tolerance; that is, the failure of T cells to respond. However, this may not generally apply. Here we report several important findings regarding the immune response to tumours, on the basis of studies of several tumour types. First, tumour-specific induction of protective cytotoxic T cells (CTLs) depends on sufficient tumour cells reaching secondary lymphatic organs early and for a long enough duration. Second, diffusely invading systemic tumours delete CTLs. Third, tumours that stay strictly outside secondary lymphatic organs, or that are within these organs but separated from T cells by barriers, are ignored by T cells but do not delete them. Fourth, co-stimulatory molecules on tumour cells do not influence CTL priming but enhance primed CTL responses in peripheral solid tumours. Last, cross priming of CTLs by tumour antigens, mediated by major histocompatibility complex (MHC) class I molecules of antigen-presenting host cells, is inefficient and not protective. These rules of T-cell induction and maintenance not only change previous views but also rationales for anti-tumour immunotherapy.


Subject(s)
Immunologic Surveillance , Lymphatic Metastasis/immunology , Lymphoid Tissue/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Antigens, Viral/immunology , B7-1 Antigen/immunology , CD28 Antigens/immunology , Glycoproteins/immunology , Histocompatibility Antigens Class I/immunology , Lymph Nodes/immunology , Lymphocyte Activation , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Signal Transduction , Transfection , Tumor Cells, Cultured , Tumor Escape/immunology
14.
J Immunol ; 166(6): 3678-87, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238607

ABSTRACT

This study evaluated to what extent presentation of exogenously acquired self-Ags via MHC class I molecules on DC might contribute to the activation of self-reactive CTL and subsequent development of autoimmune disease. We show here by using the rat insulin promotor lymphocytic choriomeningitis virus glycoprotein model of autoimmune diabetes that the activation of self-reactive CTL by DC after uptake of exogenous Ag is very limited, first by the short half-life of MHC class I-associated peptides on DC in vitro and in vivo, and second by the rather inefficient MHC class I presentation of cell-associated self-Ags by DC. These two mechanisms are probably crucial in establishing high thresholds for the induction of self-reactive CTL that prevent autoimmune sequelae after release of sequestered and previously immunologically ignored tissue Ags.


Subject(s)
Antigen Presentation , Antigens, Viral/immunology , Antigens, Viral/metabolism , Dendritic Cells/immunology , Lymphocyte Activation , Peptides/immunology , Peptides/metabolism , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins , Amino Acid Sequence , Animals , Antigen Presentation/genetics , Antigens, Viral/genetics , Cytotoxicity, Immunologic/genetics , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Glycoproteins/immunology , Glycoproteins/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Injections, Subcutaneous , Insulin/genetics , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocyte Activation/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Peptide Fragments/immunology , Peptide Fragments/metabolism , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Rats , T-Lymphocytes, Helper-Inducer/immunology , Tumor Cells, Cultured/transplantation
15.
Immunol Today ; 21(12): 624-30, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11114423

ABSTRACT

Natural or spontaneous antibodies are an essential part of the first line of defense against hematogenically spreading infections, including viruses. These antibodies target virus-antibody complexes and complement to the spleen. This prevents infections from reaching vital organs and enhances neutralizing antibody responses, particularly when the antibody is bound to a highly repetitive antigen.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Viral/immunology , Antibody Formation , Complement System Proteins/physiology , Immunity, Innate , Animals , Antigen-Antibody Complex/immunology , Antigens, Viral/immunology , Bacteremia/immunology , Bacterial Infections/immunology , Complement Activation , Germ-Free Life , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphocyte Subsets/immunology , Macrophages/immunology , Mice , Mucous Membrane/immunology , Spleen/immunology , Viremia/immunology , Virus Diseases/immunology
16.
Proc Natl Acad Sci U S A ; 97(24): 13263-8, 2000 Nov 21.
Article in English | MEDLINE | ID: mdl-11069289

ABSTRACT

Memory is a hallmark of immunity. Memory carried by antibodies is largely responsible for protection against reinfection with most known acutely lethal infectious agents and is the basis for most clinically successful vaccines. However, the nature of long-term B cell and antibody memory is still unclear. B cell memory was studied here after infection of mice with the rabies-like cytopathic vesicular stomatitis virus, the noncytopathic lymphocytic choriomeningitis virus (Armstrong and WE), and after immunization with various inert viral antigens inducing naive B cells to differentiate either to plasma cells or memory B cells in germinal centers of secondary lymphoid organs. The results show that in contrast to very low background levels against internal viral antigens, no significant neutralizing antibody memory was observed in the absence of antigen and suggest that memory B cells (i) are long-lived in the absence of antigen, nondividing, and relatively resistant to irradiation, and (ii) must be stimulated by antigen to differentiate to short-lived antibody-secreting plasma cells, a process that is also efficient in the bone marrow and always depends on radiosensitive, specific T help. Therefore, for vaccines to induce long-term protective antibody titers, they need to repeatedly provide, or continuously maintain, antigen in minimal quantities over a prolonged time period in secondary lymphoid organs or the bone marrow for sufficient numbers of long-lived memory B cells to mature to short-lived plasma cells.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory , Lymphoid Tissue/immunology , T-Lymphocytes/immunology , Vesicular stomatitis Indiana virus/immunology , Animals , Antibodies, Viral/biosynthesis , Antibody Formation , Bone Marrow Cells/immunology , Cells, Cultured , Mice , Mice, Inbred C57BL , Plasma Cells/immunology , Spleen/immunology , Time Factors , Ultraviolet Rays
17.
Mol Cell Biol ; 20(15): 5363-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10891477

ABSTRACT

CD37 is a membrane protein of the tetraspanin superfamily, which includes CD9, CD53, CD63, CD81, and CD82. Many of these molecules are expressed on leukocytes and have been implicated in signal transduction, cell-cell interactions, and cellular activation and development. We generated and analyzed mice deficient for CD37. Despite the high expression of CD37 on cells of the immune system, no changes in development and cellular composition of lymphoid organs were observed in mice lacking CD37. Analyses of humoral immune responses revealed a reduced level of immunoglobulin G1 (IgG1) in the sera of nonimmunized mice and an alteration of responses to T-cell-dependent antigens. Antibody responses to model antigen administered in the absence of adjuvant and to viral infections were generally poor in CD37-deficient mice. These poor antibody responses could be overcome by the immunization of antigen together with adjuvant. These results suggest a role for CD37 in T-cell-B-cell interactions which manifests itself under suboptimal costimulatory conditions.


Subject(s)
Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Neoplasm , B-Lymphocytes/immunology , Glycoproteins/genetics , Glycoproteins/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/physiology , Bone Marrow/immunology , Ficoll/analogs & derivatives , Ficoll/immunology , Gene Silencing , Haptens , Hemocyanins/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immunization , Immunoglobulin G/immunology , Mice , Mice, Mutant Strains , Recombination, Genetic , Rhabdoviridae Infections/immunology , Strongylida Infections/immunology , T-Lymphocytes/physiology , Tetraspanins , Th1 Cells/immunology , Th1 Cells/parasitology , Th2 Cells/immunology , Th2 Cells/parasitology , Trinitrobenzenes/immunology
18.
J Immunol ; 164(12): 6296-302, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843683

ABSTRACT

Many natural viral and bacterial pathogens activate B cells independently of Th cells (TI Ags). This study analyzed the characteristics of the activation of B cells after immunization with various forms of viral Ags using different immunization routes and found a decreasing dependence on T help with increasing amounts of Ag recruited to the spleen. Repetitive antigenic structure facilitated TI B cell responses if Ag was present in lymphoid organs. These results suggest that 1) Ag dose and localization in secondary lymphoid organs are the key for B cell activation in the absence of T help; 2) early TI Ab responses are crucial to protect against systemically spreading acute cytopathic infectious agents; and 3) there may be new rationales for improved vaccine design.


Subject(s)
Antibodies, Viral/biosynthesis , Antigens, T-Independent/physiology , Antigens, Viral/administration & dosage , Antigens, Viral/immunology , Lymphoid Tissue/immunology , Splenectomy , Viral Vaccines/immunology , Animals , Antibodies, Viral/physiology , Antigens, Viral/chemistry , Dose-Response Relationship, Immunologic , Immunohistochemistry , Injections, Intravenous , Injections, Subcutaneous , Lymphoid Tissue/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Nude , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/prevention & control , Structure-Activity Relationship , Vesicular stomatitis Indiana virus/immunology , Viral Vaccines/chemical synthesis
19.
J Immunol ; 164(11): 5761-70, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10820254

ABSTRACT

FTY720 (2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol hydrochloride) prolongs survival of solid organ allografts in animal models. Mechanisms of FTY720 immunomodulation were studied in mice infected with lymphocytic choriomeningitis virus (LCMV) to assess T cell responses or with vesicular stomatitis virus to evaluate Ab responses. Oral FTY720 (0.3 mg/kg/day) did not affect LCMV replication and specific CTL and B cells were induced and expanded normally. Moreover, the anti-viral humoral immune responses were normal. However, FTY720 treatment showed first a shift of overall distribution of CTL from the spleen to peripheral lymph nodes and lymphocytopenia was observed. This effect was reversible within 7-21 days. Together with unimpaired T and B cell memory after FTY720 treatment, this finding rendered enhancement of lymphocyte apoptosis by FTY720 in vivo unlikely. Secondly, the delayed-type hypersensitivity reaction to a viral MHC class I-presented peptide was markedly reduced by FTY720. These results were supported by impaired circulation of LCMV specific TCR transgenic effector lymphocytes in the peripheral blood and reduced numbers of tissue infiltrating CTL in response to delayed-type hypersensitivity reaction. Thirdly, in a CD8+ T cell-mediated diabetes model in a transgenic mouse expressing the LCMV glycoprotein in the islets of the pancreas, FTY720 delayed or prevented disease by reducing islet-infiltrating CTL. Thus, FTY720 effectively reduced recirculation of CD8+ effector T cells and their recruitment to peripheral lesions without affecting the induction and expansion of immune responses in secondary lymphoid organs. These properties may offer the potential to treat ongoing organ-specific T cell-mediated immunopathologic disease.


Subject(s)
Cell Movement/drug effects , Cytotoxicity, Immunologic/drug effects , Immunologic Memory/drug effects , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Propylene Glycols/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Viral/biosynthesis , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/prevention & control , Edema/blood , Edema/immunology , Edema/pathology , Edema/virology , Fingolimod Hydrochloride , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Hypersensitivity, Delayed/virology , Lymphocyte Count , Lymphocytic choriomeningitis virus/immunology , Lymphopenia/blood , Lymphopenia/immunology , Lymphopenia/pathology , Lymphopenia/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sphingosine/analogs & derivatives , T-Lymphocytes, Cytotoxic/pathology
20.
J Exp Med ; 191(5): 795-804, 2000 Mar 06.
Article in English | MEDLINE | ID: mdl-10704461

ABSTRACT

Vaccination with dendritic cells (DCs) presenting tumor antigens induces primary immune response or amplifies existing cytotoxic antitumor T cell responses. This study documents that antitumor treatment with DCs may cause severe autoimmune disease when the tumor antigens are not tumor-specific but are also expressed in peripheral nonlymphoid organs. Growing tumors with such shared tumor antigens that were, at least initially, strictly located outside of secondary lymphoid organs were successfully controlled by specific DC vaccination. However, antitumor treatment was accompanied by fatal autoimmune disease, i.e., autoimmune diabetes in transgenic mice expressing the tumor antigen also in pancreatic beta islet cells or by severe arteritis, myocarditis, and eventually dilated cardiomyopathy when arterial smooth muscle cells and cardiomyocytes expressed the transgenic tumor antigen. These results reveal the delicate balance between tumor immunity and autoimmunity and therefore point out important limitations for the use of not strictly tumor-specific antigens in antitumor vaccination with DCs.


Subject(s)
Antigens, Neoplasm/therapeutic use , Autoimmune Diseases/etiology , Cancer Vaccines/adverse effects , Dendritic Cells/immunology , Fibrosarcoma/therapy , Vaccination/adverse effects , Animals , Cardiovascular Diseases/etiology , Cardiovascular Diseases/immunology , Diabetes Mellitus, Type 1/etiology , Glycoproteins/immunology , Islets of Langerhans/immunology , Lung/pathology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Transgenic , Myocardium/pathology , Neoplasms, Experimental/therapy , Peptide Fragments/immunology , beta-Galactosidase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...