Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Immunol ; 7(73): eabm3723, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35857577

ABSTRACT

Antibodies protect from infection, underpin successful vaccines and elicit therapeutic responses in otherwise untreatable cancers and autoimmune conditions. The human IgG2 isotype displays a unique capacity to undergo disulfide shuffling in the hinge region, leading to modulation of its ability to drive target receptor signaling (agonism) in a variety of important immune receptors, through hitherto unexplained molecular mechanisms. To address the underlying process and reveal how hinge disulfide orientation affects agonistic activity, we generated a series of cysteine to serine exchange variants in the hinge region of the clinically relevant monoclonal antibody ChiLob7/4, directed against the key immune receptor CD40. We report how agonistic activity varies with disulfide pattern and is afforded by the presence of a disulfide crossover between F(ab) arms in the agonistic forms, independently of epitope, as observed in the determined crystallographic structures. This structural "switch" affects directly on antibody conformation and flexibility. Small-angle x-ray scattering and ensemble modeling demonstrated that the least flexible variants adopt the fewest conformations and evoke the highest levels of receptor agonism. This covalent change may be amenable for broad implementation to modulate receptor signaling in an epitope-independent manner in future therapeutics.


Subject(s)
Disulfides , Immunoglobulin G , Antibodies, Monoclonal , Disulfides/chemistry , Epitopes , Humans , Protein Conformation
2.
J Biol Chem ; 290(9): 5424-37, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25568316

ABSTRACT

Type I anti-CD20 mAb such as rituximab and ofatumumab engage with the inhibitory FcγR, FcγRIIb on the surface of B cells, resulting in immunoreceptor tyrosine-based inhibitory motif (ITIM) phosphorylation. Internalization of the CD20·mAb·FcγRIIb complex follows, the rate of which correlates with FcγRIIb expression. In contrast, although type II anti-CD20 mAb such as tositumomab and obinutuzumab also interact with and activate FcγRIIb, this interaction fails to augment the rate of CD20·mAb internalization, raising the question of whether ITIM phosphorylation plays any role in this process. We have assessed the molecular requirements for the internalization process and demonstrate that in contrast to internalization of IgG immune complexes, FcγRIIb-augmented internalization of rituximab-ligated CD20 occurs independently of the FcγRIIb ITIM, indicating that signaling downstream of FcγRIIb is not required. In transfected cells, activatory FcγRI, FcγRIIa, and FcγRIIIa augmented internalization of rituximab-ligated CD20 in a similar manner. However, FcγRIIa mediated a slower rate of internalization than cells expressing equivalent levels of the highly homologous FcγRIIb. The difference was maintained in cells expressing FcγRIIa and FcγRIIb lacking cytoplasmic domains and in which the transmembrane domains had been exchanged. This difference may be due to increased degradation of FcγRIIa, which traffics to lysosomes independently of rituximab. We conclude that the cytoplasmic domain of FcγR is not required for promoting internalization of rituximab-ligated CD20. Instead, we propose that FcγR provides a structural role in augmenting endocytosis that differs from that employed during the endocytosis of immune complexes.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/metabolism , Antigens, CD20/metabolism , Endocytosis , Receptors, IgG/metabolism , Biotinylation , Blotting, Western , Cell Line, Tumor , Humans , Microscopy, Confocal , Mutation , Receptors, IgG/genetics , Rituximab , Signal Transduction
3.
Blood ; 123(5): 669-77, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24227819

ABSTRACT

A major feature that distinguishes type I from type II anti-CD20 monoclonal antibodies (mAbs) and reduces their therapeutic efficacy is the tendency to internalize from the cell surface. We have shown previously that the extent of internalization correlates with the capacity of type I mAb to simultaneously engage both CD20 and the inhibitory Fcγ receptor, FcγRIIb, in a bipolar configuration. Here, we investigated whether mAbs directed at other B-cell surface receptors also engaged FcγRIIb and whether this interaction promoted internalization. Most mAbs engaged and activated FcγRIIb, with the strength of activation related to the level of mAb bound to the cell surface. However, engagement did not affect internalization of most mAb-ligated receptors, either in cell lines or primary chronic lymphocytic leukemia cells with the exception of CD19 and CD38. Furthermore, at high cell concentrations/density both cis and trans interactions between cell-surface bound mAb and FcγRIIb were evident, but trans interactions did not inhibit type I anti-CD20 mAb-mediated internalization. These data identify that FcγRIIb is engaged by many mAbs in both cis and trans configurations, triggering its activation, but that internalization via FcγRIIb occurs for only a select subset. These findings have implications when designing new antibody-based therapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Receptors, IgG/immunology , Antibodies, Monoclonal/metabolism , Antibody Specificity , Antigens, CD20/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Humans , Protein Isoforms/immunology , Protein Transport
4.
J Immunol ; 191(8): 4130-40, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24026082

ABSTRACT

Genetic deficiency of the inhibitory Fc receptor, FcγRIIB (CD32b), has been shown to augment the activity of activatory FcγR and promote mAb immunotherapy. To investigate whether mAbs capable of blocking FcγRIIB have similar capacity, we recently generated a panel of specific anti-mouse FcγRIIB mAbs that do not cross-react with other FcRs, allowing us to study the potential of FcγRIIB as a therapeutic target. Previous work revealed a number of these mAbs capable of eliciting programmed cell death of targets, and in the present study we demonstrated their ability to promote target cell phagocytosis. However, in a variety of murine tumor models, anti-FcγRIIB mAbs demonstrated limited therapeutic activity despite optimized treatment regimens. Unexpectedly, we observed that the anti-FcγRIIB mAbs are rapidly and extensively consumed in vivo, both by the tumor and host cells, including B cells, leading to a precipitous loss from the circulation. Closer analysis revealed that the anti-FcγRIIB mAbs become extensively internalized from the cell surface within 24 h in vivo, likely explaining their suboptimal efficacy. Subsequent studies revealed that anti-FcγRIIB mAb immunotherapy was effective when used against FcγRIIB(+) tumors in FcγRIIB(-/-) recipients, indicating that consumption of the mAb by nontumor cells is the primary limitation of these reagents. Importantly, similar rates of internalization were not seen on human target cells, at least in vitro. These studies further highlight the need to determine the propensity of mAb therapeutics to internalize target receptors and also identify potential key differences between human and mouse cells in this respect.


Subject(s)
Antibodies, Monoclonal/immunology , Lymphoma, B-Cell/immunology , Macrophages/immunology , Multiple Myeloma/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/metabolism , Apoptosis/immunology , Cells, Cultured , Female , Humans , Immunotherapy , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Multiple Myeloma/therapy , Receptors, IgG/genetics
5.
Blood ; 117(17): 4519-29, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21378274

ABSTRACT

The anti-CD20 mAb rituximab has substantially improved the clinical outcome of patients with a wide range of B-cell malignancies. However, many patients relapse or fail to respond to rituximab, and thus there is intense investigation into the development of novel anti-CD20 mAbs with improved therapeutic efficacy. Although Fc-FcγR interactions appear to underlie much of the therapeutic success with rituximab, certain type II anti-CD20 mAbs efficiently induce programmed cell death (PCD), whereas rituximab-like type I anti-CD20 mAbs do not. Here, we show that the humanized, glycoengineered anti-CD20 mAb GA101 and derivatives harboring non-glycoengineered Fc regions are type II mAb that trigger nonapoptotic PCD in a range of B-lymphoma cell lines and primary B-cell malignancies. We demonstrate that GA101-induced cell death is dependent on actin reorganization, can be abrogated by inhibitors of actin polymerization, and is independent of BCL-2 overexpression and caspase activation. GA101-induced PCD is executed by lysosomes which disperse their contents into the cytoplasm and surrounding environment. Taken together, these findings reveal that GA101 is able to potently elicit actin-dependent, lysosomal cell death, which may potentially lead to improved clearance of B-cell malignancies in vivo.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Death/immunology , Lymphoma, B-Cell , Actins/drug effects , Actins/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived/pharmacology , Cathepsins/pharmacology , Cell Adhesion/immunology , Cell Line, Tumor , Cell Membrane Permeability/immunology , Drug Resistance, Neoplasm/immunology , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lysosomes/drug effects , Lysosomes/immunology , Rituximab
6.
Semin Hematol ; 47(2): 107-14, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20350657

ABSTRACT

The last decade has seen the monoclonal antibody (mAb), rituximab, transform clinical management of many non-Hodgkin lymphomas and more recently provide new opportunities for controlling autoimmune conditions, such as rheumatoid arthritis. Although not yet fully determined, the explanation for this success appears to lie with the inherent properties of its target, CD20, which allow rituximab to recruit potent cytotoxic effectors with unusual efficiency. In this review we detail the properties of CD20 that make it such an effective therapeutic target and describe how different mAbs change the membrane distribution and internalization of CD20 and have distinct modes of cytotoxic activity.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, CD20/physiology , Animals , Antibodies, Monoclonal/classification , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Antibody Specificity , Antibody-Dependent Cell Cytotoxicity , Antigen-Antibody Reactions , Antigens, CD20/chemistry , Antigens, CD20/immunology , Antigens, CD20/metabolism , Antigens, Surface/immunology , Antigens, Surface/metabolism , Antineoplastic Agents/therapeutic use , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Calcium Signaling/physiology , Cell Death , Drug Delivery Systems , Humans , Lymphoma, B-Cell/drug therapy , Mice , Mice, Transgenic , Protein Conformation , Protein Transport , Rituximab
7.
J Clin Invest ; 119(8): 2143-59, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19620786

ABSTRACT

mAbs are becoming increasingly utilized in the treatment of lymphoid disorders. Although Fc-FcgammaR interactions are thought to account for much of their therapeutic effect, this does not explain why certain mAb specificities are more potent than others. An additional effector mechanism underlying the action of some mAbs is the direct induction of cell death. Previously, we demonstrated that certain CD20-specific mAbs (which we termed type II mAbs) evoke a nonapoptotic mode of cell death that appears to be linked with the induction of homotypic adhesion. Here, we reveal that peripheral relocalization of actin is critical for the adhesion and cell death induced by both the type II CD20-specific mAb tositumomab and an HLA-DR-specific mAb in both human lymphoma cell lines and primary chronic lymphocytic leukemia cells. The cell death elicited was rapid, nonapoptotic, nonautophagic, and dependent on the integrity of plasma membrane cholesterol and activation of the V-type ATPase. This cytoplasmic cell death involved lysosomes, which swelled and then dispersed their contents, including cathepsin B, into the cytoplasm and surrounding environment. The resulting loss of plasma membrane integrity occurred independently of caspases and was not controlled by Bcl-2. These experiments provide what we believe to be new insights into the mechanisms by which 2 clinically relevant mAbs elicit cell death and show that this homotypic adhesion-related cell death occurs through a lysosome-dependent pathway.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD20/metabolism , Apoptosis/drug effects , HLA-DR Antigens/metabolism , Leukemia/drug therapy , Lymphoma/drug therapy , Lysosomes/physiology , Actins/physiology , Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Autophagy , Cell Adhesion/drug effects , Cell Communication , Cell Line , HLA-DR Antigens/immunology , Humans , Leukemia/pathology , Lymphoma/pathology , Lysosomes/drug effects , Membrane Microdomains/physiology , Microvilli/physiology
8.
Blood ; 112(10): 4170-7, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18583569

ABSTRACT

Anti-CD20 monoclonal antibodies (mAbs) are classified into type I (rituximab-like) or type II (tositumomab-like) based on their ability to redistribute CD20 molecules in the plasma membrane and activate various effector functions. To compare type I and II mAbs directly in vivo and maximize Fc effector function, we selected and engineered mAbs with the same mouse IgG(2)a isotype and assessed their B-cell depleting activity in human CD20 transgenic mice. Despite being the same isotype, having similar affinity, opsonizing activity for phagocytosis, and in vivo half-life, the type II mAb tositumomab (B1) provided substantially longer depletion of B cells from the peripheral blood compared with the type I mAb rituximab (Rit m2a), and 1F5. This difference was also evident within the secondary lymphoid organs, in particular, the spleen. Failure to engage complement did not explain the efficacy of the type II reagents because type I mAbs mutated in the Fc domain (K322A) to prevent C1q binding still did not display equivalent efficacy. These results give support for the use of type II CD20 mAbs in human B-cell diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD20/immunology , Antineoplastic Agents/pharmacology , Complement Activation/drug effects , Lymphocyte Depletion/methods , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity/genetics , Antigens, CD20/metabolism , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Complement Activation/genetics , Complement Activation/immunology , Complement C1q/immunology , Complement C1q/metabolism , Drug Evaluation, Preclinical/methods , Humans , Immunoglobulin Constant Regions/genetics , Immunoglobulin Constant Regions/immunology , Mutation, Missense , Protein Binding/genetics , Protein Binding/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Rituximab
9.
J Biol Chem ; 283(25): 16971-84, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18426802

ABSTRACT

The anti-CD20 monoclonal antibody (mAb) rituximab is now routinely used for the treatment of non-Hodgkins lymphoma and is being examined in a wide range of other B-cell disorders, such as rheumatoid arthritis. Despite intensive study, the mechanism of action still remains uncertain. In the current study, anti-CD20 mAb-induced calcium signaling was investigated. Previously, we grouped anti-CD20 mAbs into Type I (rituximab-like) and Type II (B1-like) based upon various characteristics such as their ability to induce complement activation and redistribute CD20 into detergent-insoluble membrane domains. Here we show that only Type I mAbs are capable of inducing a calcium flux in B cells and that this is tightly correlated with the expression of the B-cell antigen receptor (BCR). Inhibitor analysis revealed that the signaling cascade employed by CD20 was strikingly similar to that utilized by the BCR, with inhibitors of Syk, Src, and PI3K, but not EGTA, p38, or ERK1/2, completely ablating calcium flux. Furthermore, binding of Type I but not Type II mAbs caused direct association of CD20 with the BCR as measured by FRET and resulted in the phosphorylation of BCR-specific adaptor proteins BLNK and SLP-76. Crucially, variant Ramos cells lacking BCR expression but with unchanged CD20 expression were completely unable to induce calcium flux following ligation of CD20. Collectively, these data indicate that CD20 induces cytosolic calcium flux through its ability to associate with and "hijack" the signaling potential of the BCR.


Subject(s)
Antigens, CD20/metabolism , B-Lymphocytes/metabolism , Cytosol/metabolism , Receptors, Antigen, B-Cell/metabolism , Antibodies, Monoclonal/chemistry , Calcium/chemistry , Calcium/metabolism , Calcium Signaling , Cell Communication , Cell Separation , Chelating Agents/pharmacology , Humans , Models, Biological , Phosphorylation , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...