Common Variable Immunodeficiency in Horses is Characterized by B Cell Depletion in Primary and Secondary Lymphoid Tissues

2008 ◽  
Vol 29 (1) ◽  
pp. 107-116 ◽  
Author(s):  
M. Julia B. F. Flaminio ◽  
Rebecca L. Tallmadge ◽  
Cristina O. M. Salles-Gomes ◽  
Mary Beth Matychak
2020 ◽  
Author(s):  
Pulukool Sandhya ◽  
Abhinav Jain ◽  
Geeta Govindaraj

The evolution of COVID-19 as a global pandemic and emerging evidence from cohorts of patients have provided an immense opportunity to understand biological processes pertinent to development of the disease. The immune system is central to the development of characteristic hyperinflammation. In an attempt to understand the role of humoral immunity in COVID-19, reports encompassing patients with primary immunodeficiency (PID)(13 patients) and on B cell depletion therapies(39 patients) who had concurrent COVID-19 were reviewed. In PIDs, patients with common variable immunodeficiency had worse outcomes than patients with agammaglobulinemia. Among the patients on B cell depletion therapy, heterogenous outcome was seen. As a group, patients with multiple sclerosis had better outcomes as compared to patients with systemic autoimmunity. Further, increasing reports of autoimmunity and autoinflammatory diseases occurring in temporal relation to COVID-19 is also evidence for the pathogenic role played by B cells. B cell depletion in the setting of COVID-19 may not be harmful in all patients and in fact may be protective in some scenarios. Identification of risk factors associated with a worse outcome in patients on B cell therapy could provide a rationale for individualised management.


2009 ◽  
Vol 131 ◽  
pp. S88
Author(s):  
Nilufer Seth ◽  
Cheryl Nickerson-Nutter ◽  
Quintus Medley ◽  
Lioudmila Tchistiakova ◽  
Stephane Olland ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3980-3980 ◽  
Author(s):  
Christopher J Del Nagro ◽  
Cheng Ping Mao ◽  
Martin Brovarney ◽  
Karim Dabbagh ◽  
Herbert Birnboeck ◽  
...  

Abstract Abstract 3980 Rituximab (MabThera)® is a chimeric mouse/human monoclonal antibody recognizing the B cell specific trans-membrane protein CD20. Rituximab's CD20 specific binding facilitates the prolonged depletion of both malignant and endogenous CD20+ B cells. Rituximab is currently FDA approved for monotherapy and combinatorial therapies in the treatment of multiple forms of Non Hodgkin's Lymphoma (NHL), Chronic Lymphocytic Leukemia (CLL) and Diffuse Large B cell Lymphomas (DLBCL), as well as for the treatment of moderate to severe rheumatoid arthritis patients who are refractory to TNF antagonists (in combination with methotrexate, MTX). As with most large protein based biologics, the proto-typical route of administration of rituximab is by intravenous (IV) infusion. Following delivery to the circulation, rituximab distributes into both primary and secondary lymphoid tissues throughout the body. This distribution is essential to full target coverage of CD20+ cells and therefore critical for the efficacy in targeting those cells for depletion. For other monoclonal antibodies in other clinical settings, the conversion from IV to SC administration has resulted in an improved tolerability with less infusion-related reactions, shorter administration times, an increased patient-convenience and an improved cost-effectiveness. These advantages are anticipated for the SC administration of rituximab as well. Here we describe the results of an IV/SC pharmacokinetic/pharmacodynamic (PKPD) cynomolgus monkey study comparing the traditional IV infusion of rituximab to a novel SC dosing route of rituximab administration using an SC formulation of rituximab containing recombinant human hyaluronidase (rHuPH20). rHuPH20 transiently degrades the local interstitial matrix component, hyaluronan, at the SC injection site, acting as a permeation enhancer and allowing SC administration of high volumes. Cynomolgus monkeys were treated twice, one week apart, with the rituximab SC formulation or with IV rituximab (2 × 10 mg/kg IV or SC). CD20 target coverage and depth of distal lymph node B-cell depletion was measured 7 days later, while the prolonged target coverage and duration of peripheral blood B-cell depletion was evaluated over an extended two month period. The results indicate similar rituximab trough concentrations in serum as well as similar B-cell depletion efficacy in both peripheral blood and distal secondary lymphoid tissue after IV and SC dosing. In conclusion, the results of the study suggest that the SC versus IV dosing routes do not influence the non-clinical efficacy in this B cell depletion model given that similar trough concentrations of rituximab are reached with both administration routes. Disclosures: Del Nagro: Genentech/Roche: Employment. Mao:Roche: Employment. Dabbagh:Roche: Consultancy, Employment. Birnboeck:Roche: Consultancy, Employment. Richter:Roche: Employment.


2006 ◽  
Vol 203 (3) ◽  
pp. 743-753 ◽  
Author(s):  
Yasuhito Hamaguchi ◽  
Yan Xiu ◽  
Kazuhiro Komura ◽  
Falk Nimmerjahn ◽  
Thomas F. Tedder

CD20 monoclonal antibody (mAb) immunotherapy is effective for lymphoma and autoimmune disease. In a mouse model of immunotherapy using mouse anti–mouse CD20 mAbs, the innate monocyte network depletes B cells through immunoglobulin (Ig)G Fc receptor (FcγR)-dependent pathways with a hierarchy of IgG2a/c>IgG1/IgG2b>IgG3. To understand the molecular basis for these CD20 mAb subclass differences, B cell depletion was assessed in mice deficient or blocked for stimulatory FcγRI, FcγRIII, FcγRIV, or FcR common γ chain, or inhibitory FcγRIIB. IgG1 CD20 mAbs induced B cell depletion through preferential, if not exclusive, interactions with low-affinity FcγRIII. IgG2b CD20 mAbs interacted preferentially with intermediate affinity FcγRIV. The potency of IgG2a/c CD20 mAbs resulted from FcγRIV interactions, with potential contributions from high-affinity FcγRI. Regardless, FcγRIV could mediate IgG2a/b/c CD20 mAb–induced depletion in the absence of FcγRI and FcγRIII. In contrast, inhibitory FcγRIIB deficiency significantly increased CD20 mAb–induced B cell depletion by enhancing monocyte function. Although FcγR-dependent pathways regulated B cell depletion from lymphoid tissues, both FcγR-dependent and -independent pathways contributed to mature bone marrow and circulating B cell clearance by CD20 mAbs. Thus, isotype-specific mAb interactions with distinct FcγRs contribute significantly to the effectiveness of CD20 mAbs in vivo, which may have important clinical implications for CD20 and other mAb-based therapies.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4974-4974
Author(s):  
Marna Williams ◽  
Dongwei Li ◽  
Reina Fuji ◽  
Franklin Fuh ◽  
Saileta Prabhu ◽  
...  

Abstract Antibody-based therapeutic approaches have revolutionized the treatment of non- Hodgkin’s lymphomas (NHL) as well as other hematological malignancies. However, given the biological and clinical heterogeneity of the diseases, a large variability in clinical response has been observed. Currently, patients with refractory or relapsed lymphomas have limited therapeutic options, and therefore the need to develop effective new treatments remains urgent. Antibody drug conjugates (ADC) may prove valuable in this respect. The B cell surface antigen, CD79b, is an attractive target for this approach as it is rapidly internalized upon antibody engagement and has an appropriate expression pattern, being expressed only on normal and malignant B-cells. Anti-CD79b-MCC-DM1 is an ADC consisting of a chimeric monoclonal anti-CD79b antibody, 10D10, conjugated via a thioether linker to a cytotoxic drug, DM1. DM1, a maytansinoid derivative, is a potent tubulin polymerization inhibitor. Anti-CD79b ADCs have demonstrated strong therapeutic activity against B-cell lymphoma xenografts (Polson, 2007). To investigate the pharmacodynamics (PD), pharmacokinetics (PK), and tolerability of 10D10-MCC-DM1 in nonhuman primates, we administered 2 intravenous ~30 mg/kg doses (q 3 weeks) of 10D10-MCC-DM1 or unconjugated antibody 10D10 to cynomolgus monkeys, along with vehicle controls. 10D10-MCC-DM1 and 10D10 resulted in sustained depletion of B-cells and B cell subsets in peripheral blood, with more substantial depletion in the group given 10D10- MCC-DM1. These findings correlated well with the depletion of B-cells observed in lymphoid tissues (bone marrow, lymph nodes, and spleen). We observed an initial rapid depletion of B-cells, likely attributed to probable antibody-mediated clearance by the reticuloendothelial system. This was followed by a greater sustained B-cell depletion by the ADC, attributed to the anti-mitotic pharmacology expected following B-cell receptorbound ADC internalization. In addition, unconjugated 10D10 showed substantially less depletion than 10D10-MCC-DM1 of the CD20+CD21− B cell subset, a subgroup of B cells which is phenotypically similar to human germinal center cells. There was no substantial difference in T cells or NK cells. The PD profiles are particularly encouraging from a therapeutic perspective, as the ADC may be expected to target proliferating NHL cells in a manner similar to dividing B-cells in germinal centers. The pharmacokinetics are described by the plasma measures of total antibody, conjugate, and DM1. 10D10-MCC-DM1 was cleared slowly from the plasma with a terminal half-life of 8.98 ± 1.7 days. The apparent central volume of distribution for 10D10-MCC-DM1 was 47.7 mL/kg, which approximated the physiological plasma volume in monkeys. Modest and expected accumulation was observed following the second dose. At the similar dose level of 10D10 and 10D10-MCC-DM1, the exposure of the total antibody was similar. These PK data, when compared alongside the PD results, point to the increased effectiveness of the drug conjugate in depleting B-cells. Free DM1 concentration in plasma following administration of 10D10-MCC-DM1 exhibited a profile similar to that of 10D10-MCC-DM1 but concentrations were much lower than the corresponding 10D10-MCC-DM1 with a Cmax of 189 ± 23 ng/mL. Based on the species-invariant method, it is projected that the half-life in humans for the conjugate is approximately 19.3 ± 3.4 days. Thus the pharmacokinetic profiles of the conjugate appear to support a dosing schedule of every three weeks in clinical studies. Both 10D10-MCC-DM1 and 10D10 were well tolerated. In monkeys administered 10D10-MCC-DM1, in addition to changes in the lymphoid and hematopoietic systems (B-cell depletion), minimal axonal degeneration of the sciatic nerve was observed. Trastuzumab (Herceptin)-MCC-DM1, an ADC which has shown promising safety and efficacy profiles in clinical trials, displayed similar non-B-cell related effects in cynomolgus monkeys with dose-limiting thrombocytopenia in humans. Thus it appears that targeting B-cells by an anti-CD79b ADC does not have any additional target-independent toxicity. Taken together, these preclinical data suggest that targeting CD79b with ADCs may provide a safe and effective therapy for B-cell malignancies in humans.


Sign in / Sign up

Export Citation Format

Share Document