scholarly journals B Cells Are Required for Optimal CD4+and CD8+T Cell Tumor Immunity: Therapeutic B Cell Depletion Enhances B16 Melanoma Growth in Mice

2010 ◽  
Vol 184 (7) ◽  
pp. 4006-4016 ◽  
Author(s):  
David J. DiLillo ◽  
Koichi Yanaba ◽  
Thomas F. Tedder
2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Asuka Tanaka ◽  
Kentaro Ide ◽  
Yuka Tanaka ◽  
Masahiro Ohira ◽  
Hiroyuki Tahara ◽  
...  

AbstractPretransplant desensitization with rituximab has been applied to preformed donor-specific anti-human leukocyte antigen antibody (DSA)-positive recipients for elimination of preformed DSA. We investigated the impact of pretransplant desensitization with rituximab on anti-donor T cell responses in DSA-positive transplant recipients. To monitor the patients’ immune status, mixed lymphocyte reaction (MLR) assays were performed before and after desensitization with rituximab. Two weeks after rituximab administration, the stimulation index (SI) of anti-donor CD4+ T cells was significantly higher in the DSA-positive recipients than in the DSA-negative recipients. To investigate the mechanisms of anti-donor hyper responses of CD4+ T cells after B cell depletion, highly sensitized mice models were injected with anti-CD20 mAb to eliminate B cells. Consistent with clinical observations, the SI values of anti-donor CD4+ T cells were significantly increased after anti-CD20 mAb injection in the sensitized mice models. Adding B cells isolated from untreated sensitized mice to MLR significantly inhibited the enhancement of anti-donor CD4+ T cell response. The depletion of the CD5+ B cell subset, which exclusively included IL-10-positive cells, from the additive B cells abrogated such inhibitory effects. These findings demonstrate that IL-10+ CD5+ B cells suppress the excessive response of anti-donor CD4+ T cells responses in sensitized recipients.


2019 ◽  
Vol 11 (482) ◽  
pp. eaav1648 ◽  
Author(s):  
Rita Kansal ◽  
Noah Richardson ◽  
Indira Neeli ◽  
Saleem Khawaja ◽  
Damian Chamberlain ◽  
...  

The failure of anti-CD20 antibody (Rituximab) as therapy for lupus may be attributed to the transient and incomplete B cell depletion achieved in clinical trials. Here, using an alternative approach, we report that complete and sustained CD19+ B cell depletion is a highly effective therapy in lupus models. CD8+ T cells expressing CD19-targeted chimeric antigen receptors (CARs) persistently depleted CD19+ B cells, eliminated autoantibody production, reversed disease manifestations in target organs, and extended life spans well beyond normal in the (NZB × NZW) F1 and MRLfas/fas mouse models of lupus. CAR T cells were active for 1 year in vivo and were enriched in the CD44+CD62L+ T cell subset. Adoptively transferred splenic T cells from CAR T cell–treated mice depleted CD19+ B cells and reduced disease in naive autoimmune mice, indicating that disease control was cell-mediated. Sustained B cell depletion with CD19-targeted CAR T cell immunotherapy is a stable and effective strategy to treat murine lupus, and its effectiveness should be explored in clinical trials for lupus.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 3051-3051 ◽  
Author(s):  
Youssef Hijazi ◽  
Matthias Klinger ◽  
Andrea Schub ◽  
Benjamin Wu ◽  
Min Zhu ◽  
...  

3051 Background: Blinatumomab (AMG 103) is an investigational, bispecific, T cell engaging (BiTE) antibody targeting CD19-expressing B cells. We describe the exposure-pharmacodynamic (PD) response of blinatumomab in patients with NHL, using a quantitative pharmacology approach. Methods: In a phase 1 study, 76 patients with NHL received blinatumomab by continuous intravenous infusion (cIV) at doses of 0.5 to 90 μg/m2/d in 4- or 8-week cycles. Pharmacokinetics (PK) was determined. PD responses evaluated included lymphocytes and cytokines measured during treatment, and sum of the products of the greatest diameters of tumor size in lymph nodes (SPD) at the end of treatment. Blinatumomab concentration at steady state (Css) and the cumulative area under the concentration (AUCcum)–time curve over the period before the evaluation of SPD were used to evaluate the exposure-SPD relationship. Results: Blinatumomab showed linear PK. Early PD responses were characterized by B cell depletion, T cell redistribution, and transient cytokine release. Following cIV at doses from 0.5 to 90 μg/m2/d, B cells declined at a first-order rate with a dose-dependent rate constant, ranging from 0.16 to 1.0 h-1. Complete B cell depletion was achieved within 48 hours at doses ≥5 μg/m2/d. A dose-independent decrease in T cell counts was observed within 24 hours after dosing, and T cells returned to baseline within 2 weeks of treatment. Cytokine elevation occurred in some patients and was dose-dependent. Blinatumomab exposure-SPD relationship was best described by an inhibitory Emax model (E = E0-(Imax*C)/(IC50+C)). According to the model estimation, a 50% reduction in SPD would be achieved when Css is 2141 pg/mL and AUCcum is 1381 h*μg/L, equivalent to a blinatumomab dose of 54 µg/m2/d given over 27 days. Conclusions: B lymphocytes were completely depleted from the circulation at blinatumomab doses ≥5 μg/m2/d. Depletion was faster at higher doses. Higher blinatumomab Css and AUCcum were associated with better tumor reduction. Tissue accessibility may explain the higher dose requirement for SPD reduction versus peripheral B cell depletion. The PK/PD model has utility for the design of future studies of blinatumomab in NHL. Clinical trial information: NCT00274742.


2014 ◽  
Vol 193 (2) ◽  
pp. 746-756 ◽  
Author(s):  
Jacquelyn M. Lykken ◽  
David J. DiLillo ◽  
Eric T. Weimer ◽  
Susanne Roser-Page ◽  
Mark T. Heise ◽  
...  

2019 ◽  
Vol 71 (4) ◽  
pp. 641-650 ◽  
Author(s):  
Antoine Néel ◽  
Marie Bucchia ◽  
Mélanie Néel ◽  
Gaelle Tilly ◽  
Aurélie Caristan ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (27) ◽  
pp. 5454-5463 ◽  
Author(s):  
Scott E. James ◽  
Nural N. Orgun ◽  
Thomas F. Tedder ◽  
Mark J. Shlomchik ◽  
Michael C. Jensen ◽  
...  

Abstract We have established a model of leukemia immunotherapy using T cells expressing chimeric T-cell receptors (cTCRs) targeting the CD20 molecule expressed on normal and neoplastic B cells. After transfer into human CD20 (hCD20) transgenic mice, cTCR+ T cells showed antigen-specific delayed egress from the lungs, concomitant with T-cell deletion. Few cTCR+ T cells reached the bone marrow (BM) in hCD20 transgenic mice, precluding effectiveness against leukemia. Anti-hCD20 antibody-mediated B-cell depletion before adoptive T-cell therapy permitted egress of mouse CD20-specific cTCR+ T cells from the lungs, enhanced T-cell survival, and promoted cTCR+ T cell–dependent elimination of established mouse CD20+ leukemia. Furthermore, CD20-specific cTCR+ T cells eliminated residual B cells refractory to depletion with monoclonal antibodies. These findings suggest that combination of antibody therapy that depletes antigen-expressing normal tissues with adoptive T-cell immunotherapy enhances the ability of cTCR+ T cells to survive and control tumors.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1841-1841
Author(s):  
Gordon Ng ◽  
Thomas Spreter ◽  
Rupert Davies ◽  
Grant Wickman

Abstract Although blinatumomab is an approved treatment for Philadelphia chromosome negative relapsed or refractory (r/r) precursor-B cell ALL and is under development for r/r B cell NHL, blinatumomab has several limitations impacting fuller therapeutic utility. For instance, blinatumomab therapy requires continuous infusion due to its rapid clearance owing to its size and lack of an antibody Fc and has been associated with potentially life-threatening CNS toxicities and cytokine release syndrome (Viardot A et al, Blood 2016; Goebeler ME et al, J Clin Oncol 2016; Topp et al, Lancet Oncol 2015). In addition, blinatumomab treatment is associated with higher incidence of relapse in patients with high disease burden, and its T cell redirected killing is limited by T cell immunosuppression (e.g. PD-1/PD-L1 up-regulation [Köhnke et al, J Hematol & Oncol 2015]; Treg suppression [Duell et al, ASH abstract 2014]). ZW38 is designed to address each of these limitations and represents a best in class CD19-directed CD3 T cell engager and a novel class of bispecific antibody drug conjugate (ADC). ZW38 contains an Azymetric IgG1-like Fc that carries mutations in the CH2 domain preventing FcgR dependent ADCC and ADCP and exhibits typical IgG1-like PK in rodent studies. Transient expression in mammalian CHO cells demonstrate ZW38 can be expressed at a titre of hundreds mg/L and can be purified using conventional IgG antibody methods and resins with typical IgG step purification yields and high heterodimer purity. Additionally, ZW38 has been conjugated to a microtubule inhibitor that lacks bystander killing. ZW38 antibody paratopes have been engineered to favor T cell-B cell functional engagement and selective target B cell cytotoxicity. In vitro studies demonstrated that ZW38 binds to human CD19+ B cells with >30-fold higher affinity than to human CD3+ T cells. Similar to blinatumomab, ZW38 can redirect the killing of target cancer B cells via T cell subtypes from human PBMC and its cytotoxicity is target B cell dependent. At concentrations that result in efficacious B cell depletion, ZW38 does not overly activate T cells. ZW38 is specifically engineered to induce more 'controlled' T cell activation than blinatumomab while still mediating sufficient T cell redirected target B cell depletion. ZW38 mediates T cell activation, cytokine release, and proliferation at nanomolar potency. By design, the sufficient, lower cytokine levels necessary for B cell killing may reduce the risk of cytokine release syndrome and T cell anergy. ZW38 exhibited potent growth inhibition in a panel of different B cell ALL and NHL cancer lines including but not limited to: G2, Nalm-6, RS4-11, Daudi, SUDHL-4 and SUDHL-6. In comparison to blinatumomab, ZW38 exhibits superior target B cell depletion in in vitro co-cultures of Raji lymphoma cells and human PBMC (ZW38 depletes > 90% B cells; blinatumomab depletes 20-90% B cells depending on the PBMC donor). In addition, ZW38 was effective in killing target B cells in PBMC cultures in which PD-1 has been up-regulated, which were resistant to blinatumomab killing in this assay. The dual mechanisms of action of ZW38, redirected T cell cytotoxicity and ADC cytotoxicity, may prolong and/or boost response rates, lower the incidence of relapse, and reduce the likelihood of acquired resistance while its 'controlled' T cell activation profile may reduce the risk of life-threatening and potentially fatal neurotoxicity and CRS. Disclosures Ng: Zymeworks: Employment, Patents & Royalties. Spreter:Zymeworks: Employment, Patents & Royalties. Davies:Zymeworks Biopharmaceuticals: Employment. Wickman:Zymeworks: Employment.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2785-2785
Author(s):  
Li Guo ◽  
Rukhsana Aslam ◽  
Yajing Zhao ◽  
Edwin R. Speck ◽  
Heyu Ni ◽  
...  

Abstract Primary immune thrombocytopenia (ITP) is an autoimmune disease characterized by increased platelet destruction and/or impaired megakaryocyte production, mediated by autoreactive B cells and T cells. B cell depletion therapy by rituximab, a monoclonal human anti-CD20 antibody, has been shown effective in both anti-platelet antibody positive (B cell mediated) and negative (T cell mediated) ITP patients. Those patients responsive to rituximab therapy showed normalized CD4+ and CD8+ T cell responses (Stasi et al. Blood. 2007), however, the mechanism of T cell regulation by B cell depletion is not clear. One possibility is through normalization of CD4+ T helper cells or up-regulation of CD4+ regulatory T cells (Tregs) (Stasi et al. Blood. 2008). Another possibility is by suppression of activated conventional CD8+ T cells or the up-regulation of CD8+ Tregs. We examined the changes of both CD4+ and CD8+ T cells and Tregs (CD25highFoxp3+) after B cell depletion in vivo in our ITP mouse model. Briefly, BALB/c GPIIIa (CD61) KO mice were either given PBS (ND) or mouse monoclonal anti-CD20 antibody (B-dep, Biogen) at day -1 and day 13 (250ug/mouse, ip). Residual CD19+ B cells in peripheral blood were less than 0.1% within 24hours in the latter group. All mice were immunized by transfusions of wildtype (WT) platelets at day 0, 7, 14, and 21 (1×108/mouse, iv). At day 28, we examined the percentages of T cell subsets in the spleens of the immunized mice. B cell-depleted immune CD61 KO mice showed significantly higher percentages of both CD3+CD8+ T cells and CD8+CD25highFoxp3+ T cells (Table 1). There was no significant difference in the CD3+CD4+ and CD4+CD25highFoxp3+ T cell populations. Both ND and B-dep immune CD61 KO splenocytes showed increased cytotoxicity activity against CD61+ PU5-1.8 target cells in vitro compared with naïve CD61 KO splenocytes, indicating the activation of CD8+ T cells. To test their in vivo effect on ITP development, splenocytes were engrafted from immune mice into irradiated and AsialoGM-1 treated severe combined immunodeficient (SCID) mice at a dose of 2.5×104/mouse and the mice were monitored for weekly platelet counts. ND and in vitro B cell depleted splenocytes from immune KO mice induced persistent ITP during 3 weeks observation whereas splenocytes from B-dep immune mice did not. To further confirm the role of B cell depletion on CD8+ T cell responses, CD8+ T cells from either ND or B-dep immune CD61 KO splenocytes were purified and transferred into SCID mice at 3×104/mouse. CD4+ T cells from ND immune CD61 KO splenocytes were added at 3×104/mouse to all the SCID mice to support the CD8+ T cell survival in vivo. SCID mice received CD8+ T cells from B-dep group showed higher platelet count at Day 14. Overall, our results indicate a protective role of CD8+CD25highFoxp3+ T cells against the development of cell mediated ITP that is enhanced by B cell depleting therapy in vivo. Table 1. CD61 KO MouseSpleens CD3+CD8+(%) CD8+CD25highFoxp3+ (%) Naïve Control 9.12±0.37 0.12±0.08 Immune, ND 6.78±2.37 0.0925±0.03 Immune, B-dep 14.15±5.1 0.2367±0.11 P value (ND vs B-dep) 0.0007 0.0064 Disclosures No relevant conflicts of interest to declare.


2015 ◽  
Vol 23 (10) ◽  
pp. 1653-1662 ◽  
Author(s):  
Daniel O Villarreal ◽  
Megan C Wise ◽  
Rebekah J Siefert ◽  
Jian Yan ◽  
Laurence M Wood ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document