Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Transplantation ; 106(7): 1485-1496, 2022 07 01.
Article in English | MEDLINE | ID: mdl-34966107

ABSTRACT

BACKGROUND: Imlifidase is an immunoglobulin G (IgG)-specific protease conditionally approved in the EU for desensitization in highly sensitized crossmatch positive kidney transplant patients. Imlifidase efficiently cleaves both heavy chains of IgG in a 2-step process. However, low levels of the intermediate cleavage product, single-cleaved IgG (scIgG), may persist in the circulation. The study objective was to investigate Fc-mediated effector functions of scIgG and its potential impact on common clinical immunologic assays used to assess transplant eligibility. METHODS: Imlifidase-generated scIgG, obtained by in vitro cleavage of HLA-sensitized patient serum or selected antibodies, was investigated in different complement- and FcγR-dependent assays and models, including clinical tests used to evaluate HLA-specific antibodies. RESULTS: ScIgG had significantly reduced Fc-mediated effector function compared with intact IgG, although some degree of activity in complement- and FcγR-dependent models was still detectable. A preparation of concentrated scIgG generated from a highly HLA-sensitized individual gave rise to a positive signal in the anti-HLA IgG LABScreen, which uses anti-Fc detection, but was entirely negative in the C1qScreen. The same high-concentration HLA-binding scIgG preparation also generated positive complement-dependent cytotoxicity responses against 80%-100% of donor T and B cells, although follow-up titrations demonstrated a much lower intrinsic activity than for intact anti-HLA IgG. CONCLUSIONS: ScIgG has a significantly reduced capacity to mediate Fc-dependent effector functions. However, remaining HLA-reactive scIgG in plasma after imlifidase treatment can cause positive assay results equivalent to intact IgG in clinical assays. Therefore, complete IgG cleavage after imlifidase treatment is essential to allow correct decision-making in relation to transplant eligibility.


Subject(s)
Immunoglobulin G , Kidney Transplantation , Complement System Proteins , HLA Antigens , Humans , Immunosuppressive Agents , Kidney Transplantation/adverse effects , Receptors, IgG
2.
Am J Transplant ; 21(12): 3907-3918, 2021 12.
Article in English | MEDLINE | ID: mdl-34236770

ABSTRACT

Imlifidase is a cysteine proteinase which specifically cleaves IgG, inhibiting Fc-mediated effector function within hours of administration. Imlifidase converts a positive crossmatch to a potential donor (T cell, B cell, or both), to negative, enabling transplantation to occur between previously HLA incompatible donor-recipient pairs. To date, 39 crossmatch positive patients received imlifidase prior to a kidney transplant in four single-arm, open-label, phase 2 studies. At 3 years, for patients who were AMR+ compared to AMR-, death-censored allograft survival was 93% vs 77%, patient survival was 85% vs 94%, and mean eGFR was 49 ml/min/1.73 m2 vs 61 ml/min/1.73 m2 , respectively. The incidence of AMR was 38% with most episodes occurring within the first month post-transplantation. Sub-analysis of patients deemed highly sensitized with cPRA ≥ 99.9%, and unlikely to be transplanted who received crossmatch-positive, deceased donor transplants had similar rates of patient survival, graft survival, and eGFR but a higher rate of AMR. These data demonstrate that outcomes and safety up to 3 years in recipients of imlifidase-enabled allografts is comparable to outcomes in other highly sensitized patients undergoing HLA-incompatible transplantation. Thus, imlifidase is a potent option to facilitate transplantation among patients who have a significant immunologic barrier to successful kidney transplantation. Clinical Trial: ClinicalTrials.gov (NCT02790437), EudraCT Number: 2016-002064-13.


Subject(s)
Kidney Transplantation , Desensitization, Immunologic , Graft Rejection/etiology , Graft Survival , HLA Antigens , Histocompatibility Testing , Humans , Kidney Transplantation/adverse effects
3.
Transpl Immunol ; 68: 101436, 2021 10.
Article in English | MEDLINE | ID: mdl-34265468

ABSTRACT

AIM: The aim of this study was to investigate if human IgM is a cleavable substrate for imlifidase and to explain an observed effect in anti-HLA IgM single antigen bead (SAB) assays in sensitized patients. METHODS: Serum samples collected pre- and 24 h post-imlifidase administration from sensitized patients enrolled in a phase II trial were investigated for anti-HLA IgG and IgM using SAB assays, with and without in vitro IgG depletion using a CaptureSelect™ affinity matrix. In addition, pre-dose samples and purified human IgM samples were treated with imlifidase in vitro and evaluated by SDS-PAGE, Western blot (PE-conjugated anti-human IgM) and SAB (IgG, IgM) assays. RESULTS: By comparing the mean fluorescence intensity (MFI) of HLA-beads, pre- and post-imlifidase administration, three IgM-related patterns were observed; IgM-specific HLA-SABs with an increased MFI post-imlifidase, IgM-specific HLA-SABs with a decreased MFI post-imlifidase, and IgM-specific HLA-SABs with a marginal MFI difference between the pre- and post-imlifidase administration. These IgM signal patterns were observed despite neither purified IgM nor serum IgM could be cleaved by imlifidase. After removing IgG, the effects observed on anti-HLA IgM was largely eliminated with the biggest differences seen in patients with very high anti-HLA IgG in pre-dose samples. CONCLUSION: We demonstrate that imlifidase does not cleave human IgM, including HLA-specific IgM antibodies from highly sensitized subjects. Observed decreases of SAB-HLA IgM signals after imlifidase treatment may result from the cleavage of IgG-IgM complexes which are bound to SAB-HLA. Serum analysis of patients with high levels of anti-HLA IgG will result in a more accurate SAB-HLA IgM reading after IgG depletion.


Subject(s)
HLA Antigens , Immunoglobulin G , Graft Rejection , Humans , Immunoglobulin M , Immunosuppressive Agents , Isoantibodies
4.
Transplantation ; 105(8): 1808-1817, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33093408

ABSTRACT

BACKGROUND: Highly HLA sensitized patients have limited access to life-saving kidney transplantation because of a paucity of immunologically suitable donors. Imlifidase is a cysteine protease that cleaves IgG leading to a rapid decrease in antibody level and inhibition of IgG-mediated injury. This study investigates the efficacy and safety of imlifidase in converting a positive crossmatch test to negative, allowing highly sensitized patients to be transplanted with a living or deceased donor kidney. METHODS: This open-label, single-arm, phase 2 trial conducted at 5 transplant centers, evaluated the ability of imlifidase to create a negative crossmatch test within 24 h. Secondary endpoints included postimlifidase donor-specific antibody levels compared with predose levels, renal function, and pharmacokinetic/pharmacodynamic profiles. Safety endpoints included adverse events and immunogenicity profile. RESULTS: Of the transplanted patients, 89.5% demonstrated conversion of baseline positive crossmatch to negative within 24 h after imlifidase treatment. Donor-specific antibodies most often rebounded 3-14 d postimlifidase dose, with substantial interpatient variability. Patient survival was 100% with graft survival of 88.9% at 6 mo. With this, 38.9% had early biopsy proven antibody-mediated rejection with onset 2-19 d posttransplantation. Serum IgG levels began to normalize after ~3-7 d posttransplantation. Antidrug antibody levels were consistent with previous studies. Seven adverse events in 6 patients were classified as possibly or probably related to treatment and were mild-moderate in severity. CONCLUSIONS: Imlifidase was well tolerated, converted positive crossmatches to negative, and enabled patients with a median calculated panel-reactive antibody of 99.83% to undergo kidney transplantation resulting in good kidney function and graft survival at 6 mo.


Subject(s)
Bacterial Proteins/therapeutic use , Desensitization, Immunologic/methods , Histocompatibility Testing , Kidney Transplantation , Adult , Bacterial Proteins/adverse effects , Female , Graft Rejection , Graft Survival , Humans , Isoantibodies/blood , Male , Middle Aged , Tissue Donors , Young Adult
5.
Mol Cancer Ther ; 16(9): 1887-1897, 2017 09.
Article in English | MEDLINE | ID: mdl-28533435

ABSTRACT

Endogenous plasma IgG sets an immunologic threshold that dictates the activity of tumor-directed therapeutic antibodies. Saturation of cellular antibody receptors by endogenous antibody limits antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Here, we show how enzymatic cleavage of IgG using the bacterial enzyme IdeS can be utilized to empty both high and low affinity Fcγ-receptors and clear the entire endogenous antibody pool. Using in vitro models, tumor animal models as well as ex vivo analysis of sera collected during a previous clinical trial with IdeS, we show how clearing of competing plasma antibody levels with IdeS unblocks cellular antibody receptors. We show that therapeutic antibodies against breast cancer (trastuzumab), colon cancer (cetuximab), and lymphomas (rituximab and alemtuzumab) can be potentiated when endogenous IgG is removed. Overall, IdeS is shown to be a potent tool to reboot the human antibody repertoire and to generate a window to preferentially load therapeutic antibodies onto effector cells and thereby create an armada of dedicated tumor-seeking immune cells. Mol Cancer Ther; 16(9); 1887-97. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Bacterial Proteins/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Bacterial Proteins/pharmacology , Cell Line, Tumor , Complement System Proteins , Disease Models, Animal , Drug Synergism , Humans , Mice , Phagocytosis/drug effects , Phagocytosis/immunology , Protein Binding/drug effects , Proteolysis , Receptors, IgG/metabolism , Xenograft Model Antitumor Assays
6.
J Immunol ; 195(12): 5592-601, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26553074

ABSTRACT

Ag binding to the BCR is a critical step in B cell development and activation, initiating a cascade of signaling events ultimately leading to proliferation, differentiation, or cell death. A bacterial enzyme, IgG-degrading enzyme of Streptococcus pyogenes (IdeS), was shown to specifically cleave IgG molecules below the hinge region of soluble IgG and when IgG is bound to Ag, resulting in one F(ab')2 molecule and one homodimeric Fc fragment. Whether IdeS could also cleave the IgG molecule when it is present in the BCR attached to the B cell membrane in a complex with CD79a and CD79b is unknown. In this article, we present human in vitro and ex vivo data showing that IdeS cleaves the IgG present in the BCR complex and very efficiently blocks Ag binding to the BCR. As a consequence of IdeS cleaving the BCR, signaling cascades downstream of the BCR are blocked, and memory B cells are temporarily silenced, preventing them from responding to antigenic stimulation and their transition into Ab-producing cells.


Subject(s)
Bacterial Proteins/metabolism , Immunoglobulin G/metabolism , Receptors, Antigen, B-Cell/metabolism , Streptococcus pyogenes/enzymology , Animals , Antigens/immunology , Cell Differentiation , Cell Line , Humans , Immunologic Memory , Immunomodulation , Lymphocyte Activation , Protein Binding , Signal Transduction
7.
PLoS One ; 10(7): e0132011, 2015.
Article in English | MEDLINE | ID: mdl-26177518

ABSTRACT

UNLABELLED: IdeS is a streptococcal protease that cleaves IgG antibodies into F(ab')2 and Fc fragments with a unique degree of specificity, thereby providing a novel treatment opportunity of IgG-driven autoimmune conditions and antibody mediated transplant rejection. Here we report the results from a first in man, double blinded and randomized study with single ascending doses of IdeS in healthy, male subjects. Twenty healthy subjects were given intravenous single ascending doses of IdeS. With impressive efficacy IdeS cleaved the entire plasma IgG-pool only minutes after dosing. IgG reached nadir 6-24 hours after dosing and then slowly recovered. The half-life of IdeS was 4.9 (±2.8) hours at 0.24 mg/kg with the main fraction eliminated during 24 hours. Already two hours after IdeS-dosing, the phagocytic capacity of IgG/IgG-fragments was reduced to background levels. Importantly, IdeS has the capacity to inactivate Fc-mediated effector function in vivo, was considered safe with no serious adverse events, and without dose limiting toxicity in this study. The complete, rapid, but temporary removal of IgG provides a new potent therapeutic opportunity in IgG-mediated pathogenic conditions. TRIAL REGISTRATION: ClinicalTrials.gov NCT01802697.


Subject(s)
Bacterial Proteins/pharmacology , Extracellular Space/chemistry , Immunoglobulin G/isolation & purification , Antigens, Bacterial/immunology , Bacterial Proteins/adverse effects , Bacterial Proteins/blood , Bacterial Proteins/pharmacokinetics , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Male , Phagocytosis/drug effects , Proteinuria/diagnosis , Proteolysis/drug effects
8.
J Neuroimmunol ; 173(1-2): 69-78, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16472873

ABSTRACT

Laquinimod is a novel oral immunomodulatory substance, which is currently developed for the treatment of multiple sclerosis (MS). The ability of laquinimod to inhibit disease development was investigated in chronic experimental autoimmune encephalomyelitis (chEAE) in IFN-beta k.o. mice and wild type mice. Laquinimod was shown to inhibit both disease development and histopathological changes in the CNS. Furthermore, laquinimod was found to be independent of endogenous IFN-beta for its effect in chEAE. When laquinimod was combined with exogenous IFN-beta, a synergistic disease inhibitory effect was seen. These findings using laquinimod in preclinical disease models for MS emphasize the potential of laquinimod in the future treatment of MS also in patients that do not respond to IFN-beta monotherapy. Furthermore, the results indicate that laquinimod may favourably be combined with IFN-beta.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interferon-beta/metabolism , Quinolones/therapeutic use , Animals , Central Nervous System/drug effects , Central Nervous System/pathology , Chronic Disease , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunohistochemistry , Interferon-beta/genetics , Interferon-beta/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction
9.
J Med Chem ; 47(8): 2075-88, 2004 Apr 08.
Article in English | MEDLINE | ID: mdl-15056005

ABSTRACT

Roquinimex-related 3-quinolinecarboxamide derivatives were prepared and evaluated for treatment of autoimmune disorders. The compounds were tested in mice for their inhibitory effects on disease development in the acute experimental autoimmune encephalomyelitis model and selected compounds in the beagle dog for induction of proinflammatory reaction. Structure-activity relationships are discussed. Compound 8c, laquinimod, showed improved potency and superior toxicological profile compared to the lead compound roquinimex (1b, Linomide) and was selected for clinical studies (currently in phase II).


Subject(s)
Adjuvants, Immunologic/chemical synthesis , Autoimmune Diseases/drug therapy , Quinolones/chemical synthesis , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Dogs , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Hydroxyquinolines/therapeutic use , Mice , Mice, Inbred C57BL , Quinolones/chemistry , Quinolones/therapeutic use , Structure-Activity Relationship
10.
J Neuroimmunol ; 130(1-2): 163-72, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12225898

ABSTRACT

A new orally active drug, laquinimod (ABR-215062), was shown to completely inhibit the development of murine acute experimental autoimmune encephalomyelitis (EAE). Furthermore, leukocyte infiltration into the central nervous system (CNS) was abolished in the laquinimod-treated animals. By direct comparison based on dose and total exposure, laquinimod was approximately 20 times more potent than the immunomodulator roquinimex. Laquinimod also had clear therapeutic effect when given after clinical onset in a chronic relapsing EAE model. It therefore represents a new orally active immunoregulatory drug without general immunosuppressive properties for the treatment of the autoimmune disease multiple sclerosis.


Subject(s)
Central Nervous System/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Hydroxyquinolines/pharmacology , Multiple Sclerosis/drug therapy , Neuroimmunomodulation/drug effects , Adjuvants, Immunologic/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/physiopathology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Neuroimmunomodulation/immunology , Quinolones , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...