Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Ann Rheum Dis ; 81(4): 524-536, 2022 04.
Article in English | MEDLINE | ID: mdl-35045965

ABSTRACT

BACKGROUND: Effective and safe therapies are needed for the treatment of patients with giant cell arteritis (GCA). Emerging as a key cytokine in inflammation, granulocyte-macrophage colony stimulating factor (GM-CSF) may play a role in promoting inflammation in GCA. OBJECTIVES: To investigate expression of GM-CSF and its receptor in arterial lesions from patients with GCA. To analyse activation of GM-CSF receptor-associated signalling pathways and expression of target genes. To evaluate the effects of blocking GM-CSF receptor α with mavrilimumab in ex vivo cultured arteries from patients with GCA. METHODS: Quantitative real time PCR, in situ RNA hybridisation, immunohistochemistry, immunofluorescence and confocal microscopy, immunoassay, western blot and ex vivo temporal artery culture. RESULTS: GM-CSF and GM-CSF receptor α mRNA and protein were increased in GCA lesions; enhanced JAK2/STAT5A expression/phosphorylation as well as increased expression of target genes CD83 and Spi1/PU.1 were observed. Treatment of ex vivo cultured GCA arteries with mavrilimumab resulted in decreased transcripts of CD3ε, CD20, CD14 and CD16 cell markers, and reduction of infiltrating CD16 and CD3ε cells was observed by immunofluorescence. Mavrilimumab reduced expression of molecules relevant to T cell activation (human leukocyte antigen-DR [HLA-DR]) and Th1 differentiation (interferon-γ), the pro-inflammatory cytokines: interleukin 6 (IL-6), tumour necrosis factor α (TNFα) and IL-1ß, as well as molecules related to vascular injury (matrix metalloprotease 9, lipid peroxidation products and inducible nitric oxide synthase [iNOS]). Mavrilimumab reduced CD34 + cells and neoangiogenesis in GCA lesions. CONCLUSION: The inhibitory effects of mavrilimumab on multiple steps in the GCA pathogenesis cascade in vitro are consistent with the clinical observation of reduced GCA flares in a phase 2 trial and support its development as a therapeutic option for patients with GCA.


Subject(s)
Giant Cell Arteritis , Antibodies, Monoclonal, Humanized , Arteries/metabolism , Arteries/pathology , Cells, Cultured , Cytokines , Giant Cell Arteritis/pathology , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Inflammation , Neovascularization, Pathologic , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor
2.
Genet Med ; 21(4): 887-895, 2019 04.
Article in English | MEDLINE | ID: mdl-30214072

ABSTRACT

PURPOSE: To investigate immune tolerance induction with transient low-dose methotrexate (TLD-MTX) initiated with recombinant human acid α-glucosidase (rhGAA), in treatment-naïve cross-reactive immunologic material (CRIM)-positive infantile-onset Pompe disease (IOPD) patients. METHODS: Newly diagnosed IOPD patients received subcutaneous or oral 0.4 mg/kg TLD-MTX for 3 cycles (3 doses/cycle) with the first 3 rhGAA infusions. Anti-rhGAA IgG titers, classified as high-sustained (HSAT; ≥51,200, ≥2 times after 6 months), sustained intermediate (SIT; ≥12,800 and <51,200 within 12 months), or low (LT; ≤6400 within 12 months), were compared with those of 37 CRIM-positive IOPD historic comparators receiving rhGAA alone. RESULTS: Fourteen IOPD TLD-MTX recipients at the median age of 3.8 months (range, 0.7-13.5 months) had a median last titer of 150 (range, 0-51,200) at median rhGAA duration ~83 weeks (range, 36-122 weeks). One IOPD patient (7.1%) developed titers in the SIT range and one patient (7.1%) developed titers in the HSAT range. Twelve of the 14 patients (85.7%) that received TLD-MTX remained LT, versus 5/37 HSAT (peak 51,200-409,600), 7/37 SIT (12,800-51,000), and 23/37 LT (200-12,800) among comparators. CONCLUSION: Results of TLD-MTX coinitiated with rhGAA are encouraging and merit a larger longitudinal study.


Subject(s)
Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/immunology , Immune Tolerance/genetics , Methotrexate/administration & dosage , Age of Onset , Cross Reactions/immunology , Enzyme Replacement Therapy , Female , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Humans , Infant , Infant, Newborn , Male , alpha-Glucosidases/administration & dosage , alpha-Glucosidases/genetics
3.
Am J Cardiol ; 119(6): 941-943, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28043397

ABSTRACT

A young woman, who presented with what appeared to be subacute pericarditis, was found to have primary angiosarcoma of the heart, a condition that is nearly always fatal regardless of the therapy.


Subject(s)
Heart Neoplasms/diagnosis , Hemangiosarcoma/diagnosis , Pericarditis/diagnosis , Diagnosis, Differential , Diagnostic Imaging , Electrocardiography , Fatal Outcome , Female , Heart Neoplasms/pathology , Heart Neoplasms/surgery , Hemangiosarcoma/pathology , Hemangiosarcoma/surgery , Humans , Pericarditis/pathology , Pericarditis/surgery , Young Adult
4.
AAPS J ; 19(2): 377-385, 2017 03.
Article in English | MEDLINE | ID: mdl-28083796

ABSTRACT

All biotherapeutics have the potential to induce an immune response. This immunological response is complex and, in addition to antibody formation, involves T cell activation and innate immune responses that could contribute to adverse effects. Integrated immunogenicity data analysis is crucial to understanding the possible clinical consequences of anti-drug antibody (ADA) responses. Because patient- and product-related factors can influence the immunogenicity of a therapeutic protein, a risk-based approach is recommended and followed by most drug developers to provide insight over the potential harm of unwanted ADA responses. This paper examines mitigation strategies currently implemented and novel under investigation approaches used by drug developers. The review describes immunomodulatory regimens used in the clinic to mitigate deleterious ADA responses to replacement therapies for deficiency syndromes, such as hemophilia A and B, and high risk classical infantile Pompe patients (e.g., cyclophosphamide, methotrexate, rituximab); novel in silico and in vitro prediction tools used to select candidates based on their immunogenicity potential (e.g., anti-CD52 antibody primary sequence and IFN beta-1a formulation); in vitro generation of tolerogenic antigen-presenting cells (APCs) to reduce ADA responses to factor VIII and IX in murine models of hemophilia; and selection of novel delivery systems to reduce in vivo ADA responses to highly immunogenic biotherapeutics (e.g., asparaginase). We conclude that mitigation strategies should be considered early in development for biotherapeutics based on our knowledge of existing clinical data for biotherapeutics and the immune response involved in the generation of these ADAs.


Subject(s)
Drug Design , Immunologic Factors/administration & dosage , Proteins/administration & dosage , Animals , Antibodies/immunology , Antibody Formation/immunology , Antigen-Presenting Cells/immunology , Disease Models, Animal , Drug Delivery Systems , Humans , Immunity, Innate/immunology , Immunologic Factors/adverse effects , Immunologic Factors/immunology , Mice , Proteins/adverse effects , Proteins/immunology , T-Lymphocytes/immunology
5.
J Immunol ; 193(8): 3947-58, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25210119

ABSTRACT

Biologic drugs, including enzyme-replacement therapies, can elicit anti-drug Abs (ADA) that may interfere with drug efficacy and impact patient safety. In an effort to control ADA, we focused on identifying regimens of immune tolerance induction that may be readily available for clinical use. Data generated in both wild-type mice and a Pompe disease mouse model demonstrate that single-cycle, low-dose methotrexate can be as effective as three cycles of methotrexate in providing a long-lived reduction in alglucosidase alfa-specific ADA. In addition, we show that methotrexate induces Ag-specific tolerance as mice generate similar Ab responses to an irrelevant Ag regardless of prior methotrexate treatment. Methotrexate-induced immune tolerance does not seem to involve cell depletion, but rather a specific expansion of IL-10- and TGF-ß-secreting B cells that express Foxp3, suggesting an induction of regulatory B cells. The mechanism of immune tolerance induction appears to be IL-10 dependent, as methotrexate does not induce immune tolerance in IL-10 knockout mice. Splenic B cells from animals that have been tolerized to alglucosidase alfa with methotrexate can transfer tolerance to naive hosts. We hypothesize that methotrexate induction treatment concomitant with initial exposure to the biotherapeutic can induce Ag-specific immune tolerance in mice through a mechanism that appears to involve the induction of regulatory B cells.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Folic Acid Antagonists/pharmacology , Immune Tolerance/drug effects , Methotrexate/pharmacology , alpha-Glucosidases/immunology , Adoptive Transfer , Animals , Antigens, CD1d/immunology , Antimetabolites, Antineoplastic/pharmacology , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/transplantation , CD5 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/metabolism
6.
J Immunol ; 189(2): 732-43, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22711884

ABSTRACT

Rabbit anti-thymocyte globulin (Thymoglobulin) effectively treats transplant rejection but induces anti-rabbit Ab responses, which limits routine readministration. Aiming to tolerize anti-rabbit responses, we coadministered a brief methotrexate regimen with a murine version of Thymoglobulin (mATG) for effects on anti-mATG Abs and cardiac allotransplantation in mice. Although both single and three courses of methotrexate could significantly inhibit anti-drug Ab titers to repeated mATG treatment, surprisingly, the single course given at the first mATG administration was most effective (>99% reduction). The transient methotrexate treatment also significantly improved pharmacokinetics and pharmacodynamics of repeated mATG administration. In the cardiac allograft model, the combination of transient mATG and methotrexate given only at the time of transplant dramatically improved allograft survival (>100 d) over either agent alone (<30 d). Anti-drug Ab titers were reduced and mATG exposure was increased which resulted in prolonged rather than enhanced mATG-mediated effects when combined with methotrexate. Moreover, methotrexate administration significantly reduced alloantibodies, suggesting that methotrexate not only decreases anti-drug Ab responses but also reduces Ab responses to multiple tissue-derived alloantigens simultaneously. These data suggest that mATG and methotrexate together can provide long-term allograft survival potentially through the induction of immune tolerance.


Subject(s)
Antilymphocyte Serum/administration & dosage , Graft Survival/immunology , Heart Transplantation/immunology , Immune Tolerance/drug effects , Immunosuppressive Agents/administration & dosage , Methotrexate/administration & dosage , Animals , Dose-Response Relationship, Immunologic , Drug Therapy, Combination , Graft Survival/drug effects , Heart Transplantation/pathology , Immune Tolerance/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...