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










Publication year range
1.
J Subst Use Addict Treat ; 164: 209434, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38866140

ABSTRACT

INTRODUCTION: People in treatment for alcohol use disorders (AUDs) can negatively impact their Concerned Others (COs). This longitudinal study examined patient and CO characteristics associated with CO outcomes. METHOD: Participants were 279 dyads of patients entering residential treatment and their CO. Outcomes were COs' mental health and quality of life. The study collected patient and CO predictors and CO outcomes at baseline and 3-, 6-, and 12-month follow-ups as part of a randomized controlled trial. In the first set of models, the analytic approach identified baseline patient predictors associated with COs' outcomes measured at baseline and follow-ups. In the second set of models, we examined whether those effects remained after adding baseline CO characteristics. RESULTS: In the first set of models, COs of older age and whose patient reported less CO-patient relationship stress had better mental health. Also, married COs and those with higher income and whose patient reported no violence in the CO-patient relationship had better quality of life. In the second set of models, COs whose patient entered treatment due to criminal justice involvement, with more social support, less reported stigma, and less use of avoidance coping had better mental health. Also, married COs, those with higher income, and less reported discrimination stigma had better quality of life. CONCLUSIONS: Understanding patient and CO characteristics that are associated with COs' outcomes may inform AUD treatment programs' efforts to help COs. Identifying modifiable determinants of CO outcomes is important to clinical practice regardless of whether the patient chooses to obtain treatment.

2.
Nat Commun ; 11(1): 798, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034142

ABSTRACT

Multiple myeloma (MM) is a plasma cell malignancy and most patients eventually succumb to the disease. Chimeric antigen receptor (CAR) T cells targeting B-Cell Maturation Antigen (BCMA) on MM cells have shown high-response rates, but limited durability. CD229/LY9 is a cell surface receptor present on B and T lymphocytes that is universally and strongly expressed on MM plasma cells. Here, we develop CD229 CAR T cells that are highly active in vitro and in vivo against MM plasma cells, memory B cells, and MM-propagating cells. We do not observe fratricide during CD229 CAR T cell production, as CD229 is downregulated in T cells during activation. In addition, while CD229 CAR T cells target normal CD229high T cells, they spare functional CD229neg/low T cells. These findings indicate that CD229 CAR T cells may be an effective treatment for patients with MM.


Subject(s)
Immunotherapy, Adoptive/methods , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , Antibodies/immunology , B-Lymphocytes/metabolism , Humans , K562 Cells/immunology , Male , Mice, Inbred NOD , Multiple Myeloma/pathology , Receptors, Antigen, T-Cell/metabolism , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays
4.
Oncology (Williston Park) ; 31(1): 55-63, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28090624

ABSTRACT

The introduction of cellular immunotherapies using genetically modified T cells has revolutionized the treatment of patients with B-cell lymphomas. However, despite the progress made in this field, similarly effective immunotherapeutic approaches have not yet been identified for patients with solid tumors or other hematologic malignancies such as multiple myeloma. Here we outline the most promising novel cellular immune strategies for patients with multiple myeloma. In addition, we highlight combinatorial approaches that, it is hoped, will further optimize cellular immunotherapies for myeloma and lead to deep and durable responses and, possibly, even cures.


Subject(s)
Immunotherapy/methods , Multiple Myeloma/therapy , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Multiple Myeloma/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
5.
BMC Pulm Med ; 16(1): 145, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27835955

ABSTRACT

BACKGROUND: Exercise training positively influences exercise tolerance and functional capacity of patients with idiopathic pulmonary arterial hypertension (IPAH). However, the underlying mechanisms are unclear. We hypothesized that exercise modulates the activated inflammatory state found in IPAH patients. METHODS: Single cardiopulmonary exercise testing was performed in 16 IPAH patients and 10 healthy subjects. Phenotypic characterization of peripheral blood mononuclear cells and circulating cytokines were assessed before, directly after and 1 h after exercise. RESULTS: Before exercise testing, IPAH patients showed elevated Th2 lymphocytes, regulatory T lymphocytes, IL-6, and TNF-alpha, whilst Th1/Th17 lymphocytes and IL-4 were reduced. In IPAH patients but not in healthy subject, exercise caused an immediate relative decrease of Th17 lymphocytes and a sustained reduction of IL-1-beta and IL-6. The higher the decrease of IL-6 the higher was the peak oxygen consumption of IPAH patients. CONCLUSIONS: Exercise seems to be safe from an immune and inflammatory point of view in IPAH patients. Our results demonstrate that exercise does not aggravate the inflammatory state and seems to elicit an immune-modulating effect in IPAH patients.


Subject(s)
Exercise/physiology , Familial Primary Pulmonary Hypertension/therapy , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Th17 Cells/immunology , Adult , Aged , Case-Control Studies , Exercise Tolerance/physiology , Familial Primary Pulmonary Hypertension/immunology , Female , Germany , Humans , Male , Middle Aged , Oxygen Consumption , Tumor Necrosis Factor-alpha/metabolism , Walk Test
6.
Br J Haematol ; 171(5): 752-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26456863

ABSTRACT

Cancer-testis antigens belonging to the MAGE family of genes, such as MAGEC2, are commonly and specifically expressed in Multiple Myeloma (MM) and are associated with a more aggressive clinical course and chemotherapy resistance. MAGEC2 is thought to be an excellent candidate for cancer immunotherapy; however, the biological role of MAGEC2 in MM has remained unclear. We investigated the biological role of MAGEC2 in myeloma cells determining the effect of MAGEC2 knockdown on proliferation and apoptosis. Loss of MAGEC2 resulted in reduced proliferation, viability, and anchorage-independent growth of myeloma cells irrespective of the functional status of TP53 (p53). The anti-proliferative effect of MAGEC2 silencing was due to a decrease of cells in the S phase, cell cycle delay at both G0/G1 and/or G2/M, and an increase in the sub-G0/G1 diploid population related to apoptotic cell death. Importantly, overexpression of short hairpin (sh)RNA-refractory MAGEC2 rescued the anti-proliferative effect of mRNA knockdown and protected cells from apoptotic cell death. Our findings support a TP53-independent role of MAGEC2 in promoting the survival of myeloma cells suggesting that MAGEC2-specific immunotherapies have the potential to eradicate the most malignant cells within the myeloma tumour bulk leading to durable clinical responses.


Subject(s)
Antigens, Neoplasm/physiology , Apoptosis/physiology , Multiple Myeloma/pathology , Neoplasm Proteins/physiology , Antigens, Neoplasm/genetics , Cell Cycle/genetics , Cell Enlargement , Cell Line, Tumor , Cell Proliferation/physiology , Cell Survival/genetics , Gene Knockdown Techniques , Humans , Mutation, Missense/genetics , Neoplasm Proteins/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Tumor Suppressor Protein p53/physiology
7.
J Transl Med ; 13: 197, 2015 Jun 20.
Article in English | MEDLINE | ID: mdl-26088750

ABSTRACT

BACKGROUND: Most patients with multiple myeloma (MM) will relapse after an initial response and eventually succumb to their disease. This is due to the persistence of chemotherapy-resistant tumor cells in the patients' bone marrow (BM) and immunotherapeutic approaches could contribute to eradicating these remaining cells. We evaluated SLLP1 as a potential immunotherapeutic target for MM. METHODS: We determined SLLP1 expression in myeloma cell lines and 394 BM samples from myeloma patients (n = 177) and BM samples from healthy donors (n = 11). 896 blood samples and 64 BM samples from myeloma patients (n = 263) and blood from healthy donors (n = 112) were analyzed for anti-SLLP1 antibodies. Seropositive patients were evaluated regarding SLLP1-specific T cells. RESULTS: Most cell lines showed SLLP1 RNA and protein expression while it was absent from normal BM. Of 177 patients 41% evidenced SLLP1 expression at least once during the course of their disease and 44% of newly diagnosed patients were SLLP1-positive. Expression of SLLP1 was associated with adverse cytogenetics and with negative prognostic factors including the patient's age, number of BM-infiltrating plasma cells, serum albumin, ß2-microglobulin, creatinine, and hemoglobin. Among patients treated with allogeneic stem cell transplantation those with SLLP1 expression showed a trend towards a reduced overall survival. Spontaneous anti-SLLP humoral immunity was detectable in 9.5% of patients but none of the seropositive patients evidenced SLLP1-specific T cells. However, antigen-specific T cells could readily be induced in vitro after stimulation with SLLP1. CONCLUSIONS: SLLP1 represents a promising target for the immunotherapy of MM, in particular for the adoptive transfer of T cell receptor-transduced T cells.


Subject(s)
Immunotherapy , Isoantigens/metabolism , Molecular Targeted Therapy , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Seminal Plasma Proteins/metabolism , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Antibody Formation/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , Epitopes/immunology , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunoglobulin G/immunology , Male , Middle Aged , Molecular Sequence Data , Multiple Myeloma/pathology , Phenotype , Prognosis , Receptors, Antigen, T-Cell/chemistry , T-Lymphocytes/immunology , Transplantation, Homologous
8.
Hum Vaccin Immunother ; 11(7): 1606-11, 2015.
Article in English | MEDLINE | ID: mdl-26001047

ABSTRACT

Multiple Myeloma (MM) is a plasma cell (PC) malignancy, which despite significant therapeutic advances, is still considered incurable. This is due to the persistence of chemotherapy-resistant minimal residual disease in the patients' bone marrow (BM) after an effective induction therapy. Immunotherapies targeting surface molecules expressed on the bulk of tumor cells and the chemotherapy-resistant, myeloma-propagating cells could play a central role in this clinical setting. We recently described surface molecule CD229 as a potential therapeutic target for MM. In our current study we assessed the expression of CD229 on different PC subtypes and on cells with a myeloma-propagating phenotype in a total of 77 patients with PC dyscrasias independently at 2 different cancer centers. We found that CD229 was strongly and homogeneously overexpressed on the PC of patients with monoclonal gammopathy of undetermined significance (MGUS), smoldering myeloma, MM, and PC leukemia. CD229 was particularly overexpressed on those PC showing an abnormal phenotype such as expression of CD56. Most importantly, CD229 was also highly expressed on those cells in the patients' BM displaying the phenotype of chemotherapy-resistant and myeloma-propagating cells. In conclusion, our combined findings suggest that immunotherapies targeting CD229 will not only be effective for the bulk of tumor cells but will also help to eradicate chemotherapy-resistant cells remaining in the patients' BM after induction treatment. Hopefully, the design of CD229-specific monoclonal antibodies or chimeric antigen receptor-transduced T cells will help to achieve prolonged remissions or even cures in MM patients.


Subject(s)
Antigens, CD/immunology , Multiple Myeloma/immunology , Plasma Cells/immunology , Adult , Antigens, CD/genetics , CD56 Antigen/genetics , CD56 Antigen/immunology , Drug Resistance, Neoplasm , Female , Humans , Immunophenotyping , Monoclonal Gammopathy of Undetermined Significance/immunology , Paraproteinemias/immunology , Phenotype , Plasma Cells/cytology , Signaling Lymphocytic Activation Molecule Family
9.
Clin Infect Dis ; 59(11): 1588-92, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25214510

ABSTRACT

Progressive multifocal leukoencephalopathy is a currently untreatable infection of the brain. Here, we demonstrate in 2 patients that treatment with interleukin 7, JC polyomavirus (JCV) capsid protein VP1, and a Toll-like receptor 7 agonist used as adjuvant, was well tolerated, and showed a very favorable safety profile and unexpected efficacy that warrant further investigation.


Subject(s)
Interleukin-7/therapeutic use , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/therapy , Viral Vaccines/therapeutic use , Brain/pathology , Capsid Proteins/immunology , Humans , Immunocompromised Host , JC Virus/chemistry , Leukoencephalopathy, Progressive Multifocal/drug therapy , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/prevention & control , Magnetic Resonance Imaging , Vaccines, Synthetic/therapeutic use
10.
Cancer Immunol Immunother ; 63(11): 1151-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25078248

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is the malignancy with the most frequent expression of the highly immunogenic cancer-testis antigens (CTA), and we have performed the first analysis of longitudinal expression, immunological properties, and fine specificity of CTA-specific antibody responses in MM. METHODS: Frequency and characteristics of antibody responses against cancer-testis antigens MAGE-A3, NY-ESO-1, PRAME, and SSX-2 were analyzed using peripheral blood (N = 1094) and bone marrow (N = 200) plasma samples from 194 MM patients. RESULTS: We found that antibody responses against CTA were surprisingly rare, only 2.6 and 3.1 % of patients evidenced NY-ESO-1- and SSX-2-specific antibodies, respectively. NY-ESO-1-specific responses were observed during disease progression, while anti-SSX-2 antibodies appeared after allogeneic stem cell transplantation and persisted during clinical remission. We found that NY-ESO-1- and SSX-2-specific antibodies were both capable of activating complement and increasing CTA uptake by antigen-presenting cells. SSX-2-specific antibodies were restricted to IgG3, NY-ESO-1 responses to IgG1 and IgG3. Remarkably, NY-ESO-1-positive sera recognized various non-contiguous regions, while SSX-2-specific responses were directed against a single 6mer epitope, SSX-2(85-90). CONCLUSIONS: We conclude that primary autoantibodies against intracellular MM-specific tumor antigens SSX-2 and NY-ESO-1 are rare but functional. While their contribution to disease control still remains unclear, our data demonstrate their theoretic ability to affect cellular anti-tumor immunity by formation and uptake of mono- and polyvalent immune complexes.


Subject(s)
Antigens, Neoplasm/immunology , Autoantibodies/immunology , Hematopoietic Stem Cell Transplantation , Membrane Proteins/immunology , Multiple Myeloma/immunology , Neoplasm Proteins/immunology , Repressor Proteins/immunology , Adult , Aged , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Complement Activation , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Female , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , Male , Middle Aged , Multiple Myeloma/therapy , Real-Time Polymerase Chain Reaction , Transplantation, Homologous
11.
Eur J Immunol ; 44(4): 1200-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469975

ABSTRACT

Interferon-gamma producing CD4(+) T (Th1) cells and IL-17-producing CD4(+) T (Th17) cells are involved in the pathogenesis of several autoimmune diseases including multiple sclerosis. Therefore, the development of treatment strategies controlling the generation and expansion of these effector cells is of high interest. Frankincense, the resin from trees of the genus Boswellia, and particularly its prominent bioactive compound acetyl-11-keto-ß-boswellic acid (AKBA), have potent anti-inflammatory properties. Here, we demonstrate that AKBA is able to reduce the differentiation of human CD4(+) T cells to Th17 cells, while slightly increasing Th2- and Treg-cell differentiation. Furthermore, AKBA reduces the IL-1ß-triggered IL-17A release of memory Th17 cells. AKBA may affect IL-1ß signaling by preventing IL-1 receptor-associated kinase 1 phosphorylation and subsequently decreasing STAT3 phosphorylation at Ser727, which is required for Th17-cell differentiation. The effects of AKBA on Th17 differentiation and IL-17A release make the compound a good candidate for potential treatment of Th17-driven diseases.


Subject(s)
Cell Differentiation/drug effects , Interleukin-1 Receptor-Associated Kinases/immunology , Interleukin-1beta/pharmacology , Signal Transduction/drug effects , Th17 Cells/immunology , Triterpenes/pharmacology , Adult , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Phosphorylation/drug effects , Phosphorylation/immunology , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Young Adult
12.
Hum Vaccin Immunother ; 9(12): 2533-42, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23955093

ABSTRACT

BACKGROUND: Patients with gastric cancer benefit from perioperative chemotherapy, however, treatment is toxic and many patients will relapse. The trifunctional antibody catumaxomab targets EpCAM on tumor cells, CD3 on T cells, and the Fcγ-receptor of antigen-presenting cells. While in Europe catumaxomab is approved for treating malignant ascites, it has not been investigated in the perioperative setting and its exact immunological mode of action is unclear. METHODS: In our study, gastric cancer patients received neoadjuvant platinum-based chemotherapy, one intraoperative application of catumaxomab, and 4 postoperative doses of intraperitoneal catumaxomab. Immunomonitoring was performed in 6 patients before surgery, after completion of catumaxomab treatment, and one month later. RESULTS: Intraperitoneal application of catumaxomab caused an increased expression of activation markers on the patients' T cells. This was accompanied by a transient decrease in numbers of CXCR3(+) effector T cells with a T-helper (Th)-1 phenotype in the peripheral blood. All patients evidenced pre-existing EpCAM-specific CD4(+) and/or CD8(+) T cells. While these cells transiently disappeared from the blood stream after intraperitoneal application of catumaxomab, we detected increased numbers of peripheral EpCAM-specific cells and a modified EpCAM-specific T-cell repertoire 4 weeks after completion of treatment. Finally, catumaxomab also amplified humoral immunity to tumor antigens other than EpCAM. CONCLUSIONS: Our findings suggest that catumaxomab exerts its clinical effects by (1) activating peripheral T cells, (2) redistributing effector T cells from the blood into peripheral tissues, (3) expanding and shaping of the pre-existing EpCAM-specific T-cell repertoire, and (4) spreading of anti-tumor immunity to different tumor antigens.


Subject(s)
Antibodies, Bispecific/administration & dosage , Immunologic Factors/administration & dosage , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , T-Lymphocytes/immunology , Europe , Humans , Stomach Neoplasms/immunology , Treatment Outcome
13.
Sci Transl Med ; 5(188): 188ra75, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23740901

ABSTRACT

Multiple sclerosis (MS) is a devastating inflammatory disease of the brain and spinal cord that is thought to result from an autoimmune attack directed against antigens in the central nervous system. The aim of this first-in-man trial was to assess the feasibility, safety, and tolerability of a tolerization regimen in MS patients that uses a single infusion of autologous peripheral blood mononuclear cells chemically coupled with seven myelin peptides (MOG1-20, MOG35-55, MBP13-32, MBP83-99, MBP111-129, MBP146-170, and PLP139-154). An open-label, single-center, dose-escalation study was performed in seven relapsing-remitting and two secondary progressive MS patients who were off-treatment for standard therapies. All patients had to show T cell reactivity against at least one of the myelin peptides used in the trial. Neurological, magnetic resonance imaging, laboratory, and immunological examinations were performed to assess the safety, tolerability, and in vivo mechanisms of action of this regimen. Administration of antigen-coupled cells was feasible, had a favorable safety profile, and was well tolerated in MS patients. Patients receiving the higher doses (>1 × 10(9)) of peptide-coupled cells had a decrease in antigen-specific T cell responses after peptide-coupled cell therapy. In summary, this first-in-man clinical trial of autologous peptide-coupled cells in MS patients establishes the feasibility and indicates good tolerability and safety of this therapeutic approach.


Subject(s)
Epitopes/immunology , Immune Tolerance/immunology , Leukocytes, Mononuclear/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Myelin Sheath/immunology , Peptides/immunology , Adolescent , Adult , Blood Cell Count , Demography , Disease Progression , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/physiopathology , Young Adult
14.
J Immunol ; 189(7): 3618-30, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22942431

ABSTRACT

Virus-specific CD4(+) T cells play a central role in control of viral pathogens including JC polyoma virus (JCV) infection. JCV is a ubiquitous small DNA virus that leads to persistent infection of humans with no clinical consequences. However, under circumstances of immunocompromise, it is able to cause an opportunistic and often fatal infection of the brain called progressive multifocal leukoencephalopathy (PML). PML has emerged as a serious adverse event in multiple sclerosis patients treated with the anti-VLA-4 mAb natalizumab, which selectively inhibits cell migration across the blood-brain barrier and the gut's vascular endothelium thus compromising immune surveillance in the CNS and gut. In a multiple sclerosis patient who developed PML under natalizumab treatment and a vigorous immune response against JCV after Ab washout, we had the unique opportunity to characterize in detail JCV-specific CD4(+) T cell clones from the infected tissue during acute viral infection. The in-depth analysis of 14 brain-infiltrating, JCV-specific CD4(+) T cell clones demonstrated that these cells use an unexpectedly broad spectrum of different strategies to mount an efficient JCV-specific immune response including TCR bias, HLA cross-restriction that increases avidity and influences in vivo expansion, and a combination of Th1 and Th1-2 functional phenotypes. The level of combinatorial diversity in TCR- and HLA-peptide interactions used by brain-infiltrating, JCV-specific CD4(+) T cells has not, to our knowledge, been reported before in humans for other viral infections and confirms the exceptional plasticity that characterizes virus-specific immune responses.


Subject(s)
Brain/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Gene Rearrangement, T-Lymphocyte , HLA-D Antigens/metabolism , JC Virus/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Brain/metabolism , Brain/virology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Cell Movement/genetics , Cells, Cultured , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , HLA-D Antigens/genetics , Humans , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/pathology , Receptors, Antigen, T-Cell, alpha-beta/genetics
15.
Brain ; 134(Pt 9): 2687-702, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21908874

ABSTRACT

Progressive multi-focal leucoencephalopathy and progressive multi-focal leucoencephalopathy-immune reconstitution inflammatory syndrome are caused by infection of the central nervous system with the JC polyoma virus. Both are complications of monoclonal antibody therapy in multiple sclerosis and other autoimmune diseases. Progressive multi-focal leucoencephalopathy-immune reconstitution inflammatory syndrome can obscure the diagnosis of progressive multi-focal leucoencephalopathy and lead to severe clinical disability and possibly death. Different from progressive multi-focal leucoencephalopathy, in which demyelination results from oligodendrocyte lysis by JC virus in the absence of an immune response, tissue destruction in progressive multi-focal leucoencephalopathy-immune reconstitution inflammatory syndrome is caused by a vigorous immune response within the brain. The cells and mediators that are involved in progressive multi-focal leucoencephalopathy-immune reconstitution inflammatory syndrome are as yet poorly understood. We examined two patients with multiple sclerosis, who developed progressive multi-focal leucoencephalopathy and later progressive multi-focal leucoencephalopathy-immune reconstitution inflammatory syndrome under natalizumab therapy. Due to initially negative JC viral deoxyribonucleic acid testing in the cerebrospinal fluid, a diagnostic brain biopsy was performed in one patient. Histopathology revealed brain inflammation characterized by a prominent T cell infiltrate (CD4(+)> CD8(+) T cells), but also B/plasma cells and monocytes. Despite very low JC viral load, both patients showed high intrathecal anti-JC virus antibodies. Brain-infiltrating CD4(+) T cells were studied regarding antigen specificity and function. CD4(+) T cells were highly specific for peptides from several JC virus proteins, particularly the major capsid protein VP1. T cell phenotyping revealed CD4(+) Th1 and bifunctional Th1-2 cells. The latter secrete large amounts of interferon-γ and interleukin-4 explaining the strong brain inflammation, presence of plasma cells and secretion of intrathecal anti-VP1 antibodies. The functional phenotype of brain-infiltrating JC virus-specific CD4(+) T cells was confirmed and extended by examining brain-derived JC virus-specific CD4(+) T cell clones. Our data provide novel insight into the pathogenesis of progressive multi-focal leucoencephalopathy-immune reconstitution inflammatory syndrome and indicate that JC virus-specific CD4(+) T cells play an important role in both eliminating JC virus from the brain, but also in causing the massive inflammation with often fatal outcome.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Immune Reconstitution Inflammatory Syndrome/immunology , Immune Reconstitution Inflammatory Syndrome/virology , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/immunology , Adult , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Brain/cytology , Brain/immunology , Diagnosis, Differential , Humans , Immune Reconstitution Inflammatory Syndrome/drug therapy , Immune Reconstitution Inflammatory Syndrome/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukoencephalopathy, Progressive Multifocal/drug therapy , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Male , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Natalizumab , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...