Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Blood ; 115(11): 2283-91, 2010 Mar 18.
Article in English | MEDLINE | ID: mdl-20068220

ABSTRACT

The clinical hallmark of paroxysmal nocturnal hemoglobinuria (PNH) is chronic intravascular hemolysis that is a consequence of unregulated activation of the alternative pathway of complement (APC). Intravascular hemolysis can be inhibited in patients by treatment with eculizumab, a monoclonal antibody that binds complement C5 thereby preventing formation of the cytolytic membrane attack complex of complement. However, in essentially all patients treated with eculizumab, persistent anemia, reticulocytosis, and biochemical evidence of hemolysis are observed; and in a significant proportion, their PNH erythrocytes become opsonized with complement C3. These observations suggest that PNH patients treated with eculizumab are left with clinically significant immune-mediated hemolytic anemia because the antibody does not block APC activation. With a goal of improving PNH therapy, we characterized the activity of anti-C3b/iC3b monoclonal antibody 3E7 in an in vitro model of APC-mediated hemolysis. We show that 3E7 and its chimeric-deimmunized derivative H17 block both hemolysis and C3 deposition on PNH erythrocytes. The antibody is specific for the APC C3/C5 convertase because classical pathway-mediated hemolysis is unaffected by 3E7/H17. These findings suggest an approach to PNH treatment in which both intravascular and extravascular hemolysis can be inhibited while preserving important immune functions of the classical pathway of complement.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody Specificity/drug effects , Complement C3/immunology , Complement Pathway, Alternative/drug effects , Hemoglobinuria, Paroxysmal/immunology , Hemoglobinuria, Paroxysmal/prevention & control , Hemolysis/drug effects , Acids , Antibodies, Monoclonal/therapeutic use , Complement C5/deficiency , Cytotoxicity, Immunologic/drug effects , Erythrocytes/drug effects , Erythrocytes/immunology , Erythrocytes/pathology , Hemoglobinuria, Paroxysmal/drug therapy , Humans , Models, Immunological , Protein Binding/drug effects
2.
Haematologica ; 95(2): 329-32, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19679883

ABSTRACT

A pilot study previously demonstrated that thrice-weekly, fractionated-dose intravenous rituximab (RTX) limits CD20 loss from chronic lymphocytic leukemia (CLL) B cells, thereby enhancing immunotherapeutic targeting. Here, we investigated the feasibility of giving 20 mg rituximab subcutaneously thrice weekly for up to 12 weeks in 4 previously treated CLL patients. Subcutaneous rituximab was well-tolerated with minimal injection site reactions; a variable degree of efficacy was observed, likely influenced by the size of the patients' B cell/CD20 burden. Subcutaneous RTX largely preserved CD20 expression on leukemic cells but the most effective therapeutic dosing regimen needs to be established (ClinicalTrials.gov Identifier: NCT00366418).


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD20/genetics , Antineoplastic Agents/therapeutic use , Gene Expression/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Lymphocyte Count , Male , Middle Aged , Rituximab
3.
J Immunol ; 183(1): 749-58, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19535640

ABSTRACT

The CD20 mAb ofatumumab (OFA) is more effective than rituximab (RTX) in promoting complement-dependent cytotoxicity (CDC) of B cells via the classical pathway (CP) of complement. CP activation is initiated by C1q binding to cell-bound IgG. Therefore, we examined the role of C1q in the dynamics of complement activation and CDC of B cell lines and primary cells from patients with chronic lymphocytic leukemia, reacted with OFA or RTX. C1q binding, complement activation, and colocalization of C1q with cell-bound mAbs were determined by flow cytometry and high-resolution digital imaging. C1q binds avidly to OFA-opsonized Raji and Daudi cells (K(D) = 12-16 nM) and colocalizes substantially with cell-bound OFA. Cells opsonized with OFA undergo high levels of complement activation and CDC in C1q-depleted serum supplemented with low concentrations of C1q. Under comparable conditions, RTX-opsonized cells bind less C1q; in addition, even when higher concentrations of C1q are used to achieve comparable C1q binding to RTX-opsonized cells, less complement activation and CDC are observed. Greater CDC induced by OFA may occur because C1q is bound in close proximity and with high avidity to OFA, resulting in effective CP activation. Moreover, OFA binds to the small, extracellular CD20 loop, placing the mAb considerably closer to the cell membrane than does RTX. This may facilitate effective capture and concentration of activated complement components closer to the cell membrane, potentially shielding them from inactivation by fluid phase agents and promoting efficient generation of the membrane attack complex.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigens, CD20/immunology , B-Lymphocyte Subsets/immunology , Complement C1q/metabolism , Cytotoxicity Tests, Immunologic , Opsonin Proteins/metabolism , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , B-Lymphocyte Subsets/metabolism , Cell Line, Tumor , Complement C1q/physiology , Complement C3b/metabolism , Complement Pathway, Classical/immunology , Cytotoxicity Tests, Immunologic/methods , Dose-Response Relationship, Immunologic , Humans , Protein Binding/immunology , Rituximab
4.
J Immunol ; 181(11): 8120-32, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19018005

ABSTRACT

More than 20 years ago clinical investigations in the immunotherapy of cancer revealed that infusion of certain immunotherapeutic mAbs directed to tumor cells induced loss of targeted epitopes. This phenomenon, called antigenic modulation, can compromise mAb-based therapies. Recently we reported that rituximab (RTX) treatment of chronic lymphocytic leukemia patients induced substantial loss of targeted CD20 on B cells found in the circulation after RTX infusion; this "shaving" of RTX-CD20 complexes from B cells is also promoted in vitro by THP-1 monocytes and by PBMC in a reaction mediated by Fcgamma receptors. The mechanism responsible for shaving appears to be trogocytosis, a process in which receptors on effector cells remove and internalize cognate ligands and cell membrane fragments from target cells. We now report that three therapeutic mAbs approved by the U.S. Food and Drug Administration for the treatment of cancer, RTX, cetuximab, and trastuzumab, as well as mAb T101, which has been shown to induce antigenic modulation in the clinic, promote trogocytosis in vitro upon binding to their respective target cells. Trogocytosis of the mAb-opsonized cells is mediated by THP-1 monocytes and by primary monocytes isolated from PBMC. In view of these results, it is likely that these mAbs and possibly other anticancer mAbs now used in the clinic may promote trogocytic removal of the therapeutic mAbs and their cognate Ags from tumor cells in vivo. Our findings may have important implications with respect to the use of mAbs in cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , Antirheumatic Agents/immunology , B-Lymphocytes/immunology , Neoplasms/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20/immunology , Antigens, Neoplasm/immunology , Antirheumatic Agents/pharmacology , Antirheumatic Agents/therapeutic use , Cell Line, Tumor , Cetuximab , Humans , Mice , Monocytes , Neoplasms/drug therapy , Receptors, IgG/immunology , Rituximab , Trastuzumab
5.
J Immunol ; 181(1): 822-32, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18566448

ABSTRACT

Binding of the CD20 mAb rituximab (RTX) to B lymphocytes in normal human serum (NHS) activates complement (C) and promotes C3b deposition on or in close proximity to cell-bound RTX. Based on spinning disk confocal microscopy analyses, we report the first real-time visualization of C3b deposition and C-mediated killing of RTX-opsonized B cells. C activation by RTX-opsonized Daudi B cells induces rapid membrane blebbing and generation of long, thin structures protruding from cell surfaces, which we call streamers. Ofatumumab, a unique mAb that targets a distinct binding site (the small loop epitope) of the CD20 Ag, induces more rapid killing and streaming on Daudi cells than RTX. In contrast to RTX, ofatumumab promotes streamer formation and killing of ARH77 cells and primary B cells from patients with chronic lymphocytic leukemia. Generation of streamers requires C activation; no streaming occurs in media, NHS-EDTA, or in sera depleted of C5 or C9. Streamers can be visualized in bright field by phase imaging, and fluorescence-staining patterns indicate they contain membrane lipids and polymerized actin. Streaming also occurs if cells are reacted in medium with bee venom melittin, which penetrates cells and forms membrane pores in a manner similar to the membrane-attack complex of C. Structures similar to streamers are demonstrable when Ab-opsonized sheep erythrocytes (non-nucleated cells) are reacted with NHS. Taken together, our findings indicate that the membrane-attack complex is a key mediator of streaming. Streamer formation may, thus, represent a membrane structural change that can occur shortly before complement-induced cell death.


Subject(s)
Antibodies, Monoclonal/immunology , B-Lymphocytes/immunology , Cell Membrane/immunology , Complement System Proteins/immunology , Lymphocyte Activation/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20/immunology , Apoptosis/drug effects , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cells, Cultured , Complement System Proteins/metabolism , Cytotoxicity, Immunologic/immunology , Humans , Protein Transport , Rituximab , Time Factors
6.
J Immunol ; 179(8): 5543-52, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911641

ABSTRACT

Childhood malaria caused by Plasmodium falciparum is often characterized by severe anemia at low parasite burdens; the mechanism(s) responsible for this pathology remain to be defined. We have reported, based on clinical observations and in vitro models, that complement control proteins on erythrocytes such as CR1, the immune adherence receptor specific for C3b, may be reduced in childhood malaria, suggesting a possible role for complement in erythrocyte destruction. Intravascular lysis of iE by P. falciparum leads to release of erythrocyte breakdown products such as hemoglobin and hematin, which have inflammatory properties. In the present article, we demonstrate that in serum and in anticoagulated whole blood, moderate concentrations of hematin activate the alternative pathway of complement and promote deposition of C3 activation and breakdown products on erythrocytes. The degree of C3 fragment deposition is directly correlated with erythrocyte CR1 levels, and erythrocytes opsonized with large amounts of C3dg form rosettes with Raji cells, which express CR2, the C3dg receptor which is expressed on several types of B cells in the spleen. Thus, the reaction mediated by hematin promotes opsonization and possible clearance of the youngest (highest CR1) erythrocytes. A mAb specific for C3b, previously demonstrated to inhibit the alternative pathway of complement, completely blocks the C3 fragment deposition reaction. Use of this mAb in nonhuman primate models of malaria may provide insight into mechanisms of erythrocyte destruction and thus aid in the development of targeted therapies based on inhibiting the alternative pathway of complement.


Subject(s)
Anemia/immunology , Complement C3/metabolism , Complement Pathway, Alternative/immunology , Erythrocytes/metabolism , Hemin/physiology , Malaria/immunology , Peptide Fragments/blood , Anemia/blood , Cell Line, Tumor , Erythrocytes/immunology , Humans , Malaria/blood , Peptide Fragments/metabolism , Protein Binding/immunology , Receptors, Complement 3b/blood , Receptors, Complement 3b/physiology , Serum
7.
J Immunol ; 179(6): 4263-71, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785867

ABSTRACT

Infusion of standard-dose rituximab (RTX) in chronic lymphocytic leukemia (CLL) patients promotes rapid complement activation and deposition of C3 fragments on CLL B cells. However, immediately after RTX infusions, there is substantial loss (shaving) of CD20 from circulating malignant cells. Because shaving can compromise efficacies of anticancer immunotherapeutic mAbs, we investigated whether shaving occurs in SCID mouse models. Z138 cells, a B cell line derived from human mantle cell lymphoma, were infused i.v. or s.c. The i.v. model recapitulates findings we previously reported for therapeutic RTX in CLL: i.v. infused RTX rapidly binds to Z138 cells in lungs, and binding is accompanied by deposition of C3 fragments. However, within 1 h targeted cells lose bound RTX and CD20, and these shaved cells are still demonstrable 40 h after RTX infusion. Z138 cells grow in tumors at s.c. injection sites, and infusion of large amounts of RTX (0.50 mg on each of 4 days) leads to considerable loss of CD20 from these cells. Human i.v. Ig blocked shaving, suggesting that FcgammaRI on cells of the mononuclear phagocytic system promote shaving. Examination of frozen tumor sections from treated mice by immunofluorescence revealed large areas of B cells devoid of CD20, with CD20 intact in adjacent areas; it is likely that RTX had opsonized Z138 cells closest to capillaries, and these cells were shaved by monocyte/macrophages. The shaving reaction occurs in neoplastic B cells in tissue and in peripheral blood, and strategies to enhance therapeutic targeting and block shaving are under development.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigens, CD/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Cell Line, Tumor , Complement C3/metabolism , Disease Models, Animal , Female , Injections, Subcutaneous , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lymphocyte Count , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/therapy , Mice , Mice, Knockout , Mice, SCID , Neoplasm Transplantation , Peptide Fragments/metabolism , Rituximab
8.
J Immunol ; 177(10): 7435-43, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082663

ABSTRACT

Treatment of chronic lymphocytic leukemia (CLL) patients with standard dose infusion of rituximab (RTX), 375 mg/m2, induces clearance of malignant cells from peripheral blood after infusion of 30 mg of RTX. After completion of the full RTX infusion, substantial recrudescence of CLL cells occurs, and these cells have lost > 90% of CD20. To gain insight into mechanism(s) of CD20 loss, we investigated the hypothesis that thrice-weekly low-dose RTX (20 or 60 mg/m2) treatment for CLL over 4 wk would preserve CD20 and enhance leukemic cell clearance. During initial infusions in all 12 patients, the first 30 mg of RTX promoted clearance of > 75% leukemic cells. Four of six patients receiving 20 mg/m2 RTX retained > or = 50% CD20, and additional RTX infusions promoted further cell clearance. However, four of six patients receiving 60 mg/m2 had CD20 levels < 20% baseline 2 days after initial infusions, and additional RTX infusions were less effective, presumably due to epitope loss. Our results suggest that when a threshold RTX dose is exceeded, recrudesced RTX-opsonized cells are not cleared, due to saturation of the mononuclear phagocytic system, but instead are shaved of RTX-CD20 complexes by acceptor cells. Thrice-weekly low-dose RTX may promote enhanced clearance of circulating CLL cells by preserving CD20.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, CD20/blood , Drug Delivery Systems , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Administration, Oral , Adult , Aged , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Complement C3/metabolism , Dose-Response Relationship, Immunologic , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Lymphocyte Count , Lymphocyte Depletion , Male , Middle Aged , Rituximab
9.
Mol Immunol ; 43(7): 1010-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-15961157

ABSTRACT

The alternative pathway (AP) of the complement system plays an important role in tissue damage and inflammation associated with certain autoimmune diseases and with ischemia-reperfusion injury. Selective inhibition of the AP could prevent such pathologies while allowing the classical and lectin pathways of complement activation to continue to provide protection. Here we present data describing selective inhibition of the AP of complement by anti-C3b/iC3b monoclonal antibody (mAb) 3E7, and by a chimeric, "deimmunized" form of this mAb, H17, which contains the human IgG1 Fc region and was further modified by substitution of amino acids in order to remove T cell epitopes. Both mAbs block AP-mediated deposition of C3b onto zymosan or Sepharose 4B, and they also inhibit AP-promoted lysis of rabbit erythrocytes. MAbs 3E7 and H17 also successfully compete with both factors B and H for binding to C3b-opsonized substrates, and the ability of both mAbs to inhibit the AP is blocked by pre-incubation with two different sources of C3(H2O). Kinetic measurements demonstrate that mAb 3E7 effectively stops progression of C3b deposition after AP activation is initiated. Our results therefore suggest that these mAbs block activation of the AP by binding to both C3(H2O) and to C3b, and thus prevent binding and activation of factor B. Based on these and other observations, mAb H17 may find future use in therapeutic applications focused on selective inhibition of the AP.


Subject(s)
Antibodies, Monoclonal/pharmacology , Complement C3b/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Antibodies, Monoclonal/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Complement Factor B/antagonists & inhibitors , Complement Factor H/antagonists & inhibitors , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology
10.
J Immunol Methods ; 294(1-2): 37-42, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15604014

ABSTRACT

Binding of rituximab (RTX) to CD20+ B cells activates complement and promotes covalent deposition of C3b fragments on the cells. Previously, we reported that the deposited C3b is substantially co-localized with cell-bound RTX, and therefore C3b may block access of antibody probes specific for RTX. We examined the ability of several commercially available phycoerythrin (PE)-labeled anti-Mouse IgG antibodies to bind to B cells opsonized in milieu which allow or preclude complement activation. Even when large amounts of fluorescently labeled RTX are bound to the cells, binding of the anti-Mouse IgG probes is substantially inhibited if C3b is deposited on the cells. However, cell-bound RTX is still demonstrable on development with a monoclonal antibody (mAb) specific for the human Fc region of RTX. Our findings may provide an alternative explanation for data presented in recent reports suggesting that binding of RTX to cells in plasma leads to internalization of RTX and CD20.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/immunology , Antigens, CD20/immunology , B-Lymphocytes/immunology , Complement C3b/immunology , Phycoerythrin/chemistry , Animals , Antibodies, Anti-Idiotypic/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20/chemistry , Antigens, CD20/metabolism , B-Lymphocytes/chemistry , Binding Sites, Antibody/immunology , Cell Line, Tumor , Complement Activation/immunology , Complement C3b/chemistry , Complement C3b/metabolism , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/immunology , Mice , Phycoerythrin/immunology , Protein Binding/immunology , Rituximab
SELECTION OF CITATIONS
SEARCH DETAIL
...