scholarly journals Preclinical activity and determinants of response of the GPRC5DxCD3 bispecific antibody talquetamab in multiple myeloma

2021 ◽  
Vol 5 (8) ◽  
pp. 2196-2215
Author(s):  
Christie P. M. Verkleij ◽  
Marloes E. C. Broekmans ◽  
Mark van Duin ◽  
Kristine A. Frerichs ◽  
Rowan Kuiper ◽  
...  

Abstract Cell surface expression levels of GPRC5D, an orphan G protein–coupled receptor, are significantly higher on multiple myeloma (MM) cells, compared with normal plasma cells or other immune cells, which renders it a promising target for immunotherapeutic strategies. The novel GPRC5D-targeting T-cell redirecting bispecific antibody, talquetamab, effectively kills GPRC5D+ MM cell lines in the presence of T cells from both healthy donors or heavily pretreated MM patients. In addition, talquetamab has potent anti-MM activity in bone marrow (BM) samples from 45 patients, including those with high-risk cytogenetic aberrations. There was no difference in talquetamab-mediated killing of MM cells from newly diagnosed, daratumumab-naïve relapsed/refractory (median of 3 prior therapies), and daratumumab-refractory (median of 6 prior therapies) MM patients. Tumor cell lysis was accompanied by T-cell activation and degranulation, as well as production of pro-inflammatory cytokines. High levels of GPRC5D and high effector:target ratio were associated with improved talquetamab-mediated lysis of MM cells, whereas an increased proportion of T cells expressing PD-1 or HLA-DR, and elevated regulatory T-cell (Treg) counts were associated with suboptimal killing. In cell line experiments, addition of Tregs to effector cells decreased MM cell lysis. Direct contact with bone marrow stromal cells also impaired the efficacy of talquetamab. Combination therapy with daratumumab or pomalidomide enhanced talquetamab-mediated lysis of primary MM cells in an additive fashion. In conclusion, we show that the GPRC5D-targeting T-cell redirecting bispecific antibody talquetamab is a promising novel antimyeloma agent. These results provide the preclinical rationale for ongoing studies with talquetamab in relapsed/refractory MM.

2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 8017-8017 ◽  
Author(s):  
Ben Buelow ◽  
Duy Pham ◽  
Starlynn Clarke ◽  
Shelley Force Aldred ◽  
Kevin Dang ◽  
...  

8017 Background: Although BCMA is a plasma cell specific surface molecule attractive as an antibody target in multiple myeloma, its scarcity on the cell surface may limit the efficacy of a conventional antibody. T-cell engaging bispecific antibody approaches are highly efficacious and are particularly well suited for a membrane target with limited expression, such as BCMA. Teneobio has developed a multivalent antibody platform based on modular human VH domains, which allowed us to build T cell engaging bispecific antibodies with low and high T cell agonistic activities. Methods: UniRats were immunized with either CD3 or BCMA antigens and antigen-specific UniAbs were identified by antibody repertoire sequencing and high-throughput gene assembly, expression, and screening. High affinity binding VH sequences were selected using recombinant proteins and cells. In vitro efficacy studies included T-cell activation by cytokine- and tumor cell kill by calcein-release assays. In vivo efficacy of the molecules was evaluated in NSG mice harboring myeloma cells and human PBMCs. Results: BCMA-specific UniAbs bound plasma cells with high affinities (100-700pM) and cross-reacted with cynomolgus plasma cells. Strong and weak T cell agonists were identified that bound human T cells with high and low affinities respectively and cross-reacted with cynomolgus T cells. T cell engaging bispecifics with a strong (H929 cytotoxicity:EC50=27pM) and a weak T cell activating arm (H929 cytotoxicity: EC50=1170pM) demonstrated T-cell activation and tumor-cell cytotoxicity in vitro; bispecifics with a weak CD3 engaging arm showed markedly reduced cytokine production even at doses saturating for cytotoxicity. In viv o, BCMAxCD3 bispecific antibodies reduced tumor load and increased survival when co-administered with human PBMCs as compared to controls. Conclusions: Our results suggest that T cell engaging bispecifics with low-affinity anti-CD3 arms could be preferred for the treatment of Multiple Myeloma.


Blood ◽  
2020 ◽  
Vol 135 (15) ◽  
pp. 1232-1243 ◽  
Author(s):  
Kodandaram Pillarisetti ◽  
Suzanne Edavettal ◽  
Mark Mendonça ◽  
Yingzhe Li ◽  
Mark Tornetta ◽  
...  

Abstract T-cell–mediated approaches have shown promise in myeloma treatment. However, there are currently a limited number of specific myeloma antigens that can be targeted, and multiple myeloma (MM) remains an incurable disease. G-protein–coupled receptor class 5 member D (GPRC5D) is expressed in MM and smoldering MM patient plasma cells. Here, we demonstrate that GPRC5D protein is present on the surface of MM cells and describe JNJ-64407564, a GPRC5DxCD3 bispecific antibody that recruits CD3+ T cells to GPRC5D+ MM cells and induces killing of GPRC5D+ cells. In vitro, JNJ-64407564 induced specific cytotoxicity of GPRC5D+ cells with concomitant T-cell activation and also killed plasma cells in MM patient samples ex vivo. JNJ-64407564 can recruit T cells and induce tumor regression in GPRC5D+ MM murine models, which coincide with T-cell infiltration at the tumor site. This antibody is also able to induce cytotoxicity of patient primary MM cells from bone marrow, which is the natural site of this disease. GPRC5D is a promising surface antigen for MM immunotherapy, and JNJ-64407564 is currently being evaluated in a phase 1 clinical trial in patients with relapsed or refractory MM (NCT03399799).


Blood ◽  
1993 ◽  
Vol 82 (6) ◽  
pp. 1787-1797 ◽  
Author(s):  
M Massaia ◽  
C Attisano ◽  
S Peola ◽  
L Montacchini ◽  
P Omede ◽  
...  

Abstract We have recently shown that peripheral blood T cells of multiple myeloma (MM) patients are very susceptible to stimulation of the T-cell receptor/CD3 complex with anti-CD3 monoclonal antibodies (MoAbs). CD3 stimulation is currently under clinical investigation as a nonspecific approach to boost antitumor effector mechanisms. The aim of this study was to determine whether the hyperreactivity of MM T cells to CD3 stimulation could be exploited to generate antitumor activity. Bone marrow mononuclear cells (BMMCs) from 65 MM patients were stimulated with the anti-CD3 MoAb OKT3 and the effect of this stimulation on autologous T cells and plasma cells was evaluated. The number of CD3+ CD25+ cells on day 6 was significantly higher in MM than the controls (30 normal individuals) (P = .001). Kinetic studies showed that 3H- thymidine incorporation peaked on day 3 and that the T-cell expansion peaked on days 5 and 6. In MM, T-cell activation markedly affected the survival of autologous plasma cells; their number in OKT3-treated cultures was significantly lower than in unstimulated cultures (P < .0001). T-cell activation and plasma cell decrease were not observed when T cells were removed from BMMC preparations. MM produced significantly higher levels of interferon-gamma (P = .005) and tumor necrosis factor-beta (P = .001), but lower levels of tumor necrosis factor-alpha (P < .001) than normal individuals. Interferon-gamma only was partially involved in CD3-induced plasma cell killing. Transwell cultures showed that the main mechanism by which CD3+ CD25+ cells affected plasma cells was direct cell-to-cell contact rather than cytokines. In conclusion, T cells in MM BMMCs possess distinct features in terms of susceptibility to CD3 stimulation and cytokine production compared with normal bone marrow T cells that can be exploited to generate antiplasma cell activity.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2824-2824 ◽  
Author(s):  
François Gaudet ◽  
Jennifer F Nemeth ◽  
Ronan McDaid ◽  
Yingzhe Li ◽  
Benjamin Harman ◽  
...  

Abstract AML is a cancer of the myeloid lineage that is characterized by the accumulation of abnormal white blood cells in the bone marrow and blood. Existing therapies do not lead to cures, partially due to their inability to eliminate residual leukemic stem cells (LSCs) in the bone marrow. T-cell redirection has been shown to be an effective method of treatment for hematologic malignancies (eg, blinatumomab) and represents an attractive approach to treat AML. CD123 (α-chain of the interleukin-3 receptor) has been shown to be expressed on the surface of AML blasts and LSCs. To eradicate CD123+ cells, we developed a bispecific antibody (JNJ-63709178) using the Genmab DuoBody® technology that can bind both CD123 on tumor cells and CD3 on T cells. JNJ-63709178 is a humanized IgG4 bispecific antibody with silenced Fc function. This antibody is able to recruit T cells to CD123-expressing tumor cells and induce the killing of these tumor cells in vitro (MOLM-13, OCI-AML5 and KG-1; EC50 = 0.51-0.91 nM). In contrast, this antibody does not kill CD123- cell lines, demonstrating the specificity of cytotoxicity. Consistently, the degree of cell killing correlated with the level of T-cell activation (CD69 and CD25) and cytokine release (TGF-β and TNF-α). Control bispecific antibodies containing a null arm (viral epitope) paired with a CD123 arm (CD123xnull) or a CD3 arm (nullxCD3) did not induce cytotoxicity or T-cell activation in the assays tested. JNJ-63709178 had no effect on T-cell activation when incubated with T cells alone. In AML murine xenograft models, JNJ-63709178 was able to suppress tumor growth and induce tumor regression (MOLM-13 and KG-1, respectively) in the presence of human peripheral blood mononuclear cells (PBMCs) or T cells. Tumor regression correlated with the infiltration of T cells in the tumor and the expression of T-cell activation markers such as CD25, PD1 and TIM3. Furthermore, this antibody was able to induce the killing of primary CD123+ cancer cells from the blood of patients with AML without the need to supplement with fresh T cells (EC50 = 0.83 nM). These results indicate that JNJ-63709178 can potently and specifically kill CD123+ cancer cells in vitro, in vivo and ex vivo. Pharmacokinetic studies in cynomolgus monkeys support twice weekly dosing for human studies. JNJ-63709178 is currently being investigated in a Phase 1 clinical trial in relapsed and refractory AML (ClinicalTrials.gov ID: NCT02715011). Disclosures Gaudet: Janssen Pharmaceuticals R&D: Employment, Other: Stock options, Patents & Royalties: pending, not yet issued. Nemeth:Janssen Pharmaceuticals R&D: Employment, Other: stock, Patents & Royalties: patent pending. McDaid:Janssen Pharmaceuticals Research and Development: Employment. Li:Janssen: Employment. Harman:Janssen Pharmaceuticals R&D: Employment. Millar:Janssen Pharmaceuticals R&D: Employment, Other: stock options. Teplyakov:Janssen Pharmaceuticals R&D: Employment. Wheeler:Janssen Pharmaceuticals R&D: Employment. Luo:Janssen Pharmaceuticals R&D: Employment. Tam:Janssen Pharmaceuticals R&D: Employment, Other: stocks, Research Funding. Wu:Janssen Pharmaceuticals R&D: Employment. Chen:Janssen Pharmaceuticals R&D: Employment. Rudnick:Janssen Pharmaceuticals R&D: Employment. Chu:Janssen Pharmaceuticals R&D: Employment. Hughes:Janssen Pharmaceuticals R&D: Employment. Luistro:Janssen: Employment. Chin:Janssen: Employment. Babich:Janssen: Employment. Kalota:Janssen Pharmaceuticals R&D: Employment, Other: stock. Singh:Janssen Pharmaceuticals R&D: Employment, Other: stock options. Salvati:Janssen Pharmaceuticals R&D: Employment, Other: stock options, Patents & Royalties: patent. Elsayed:Janssen: Employment, Other: stock options. Attar:Janssen: Employment.


Blood ◽  
1993 ◽  
Vol 82 (6) ◽  
pp. 1787-1797 ◽  
Author(s):  
M Massaia ◽  
C Attisano ◽  
S Peola ◽  
L Montacchini ◽  
P Omede ◽  
...  

We have recently shown that peripheral blood T cells of multiple myeloma (MM) patients are very susceptible to stimulation of the T-cell receptor/CD3 complex with anti-CD3 monoclonal antibodies (MoAbs). CD3 stimulation is currently under clinical investigation as a nonspecific approach to boost antitumor effector mechanisms. The aim of this study was to determine whether the hyperreactivity of MM T cells to CD3 stimulation could be exploited to generate antitumor activity. Bone marrow mononuclear cells (BMMCs) from 65 MM patients were stimulated with the anti-CD3 MoAb OKT3 and the effect of this stimulation on autologous T cells and plasma cells was evaluated. The number of CD3+ CD25+ cells on day 6 was significantly higher in MM than the controls (30 normal individuals) (P = .001). Kinetic studies showed that 3H- thymidine incorporation peaked on day 3 and that the T-cell expansion peaked on days 5 and 6. In MM, T-cell activation markedly affected the survival of autologous plasma cells; their number in OKT3-treated cultures was significantly lower than in unstimulated cultures (P < .0001). T-cell activation and plasma cell decrease were not observed when T cells were removed from BMMC preparations. MM produced significantly higher levels of interferon-gamma (P = .005) and tumor necrosis factor-beta (P = .001), but lower levels of tumor necrosis factor-alpha (P < .001) than normal individuals. Interferon-gamma only was partially involved in CD3-induced plasma cell killing. Transwell cultures showed that the main mechanism by which CD3+ CD25+ cells affected plasma cells was direct cell-to-cell contact rather than cytokines. In conclusion, T cells in MM BMMCs possess distinct features in terms of susceptibility to CD3 stimulation and cytokine production compared with normal bone marrow T cells that can be exploited to generate antiplasma cell activity.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1141-1141
Author(s):  
Elena E. Solomou ◽  
Valeria Visconte ◽  
Federica Gibellini ◽  
Neal S. Young

Abstract Ligation of the signaling lymphocyte activation molecule (SLAM), a member of the immunoglobulin superfamily expressed in T and B cells, results in T cell activation and Th1 cytokine production. SAP is a small cytoplasmic protein expressed in T- and NK cells that controls the activation signals mediated by SLAM. On T cell activation, SAP binds to Fyn kinase; Fyn is activated and phosphorylates tyrosine residues on SLAM; phosphorylation results in the formation of a complex that selectively down-regulates co-stimulatory signals in activated T cells, resulting in inhibition of IFN-γ production. Thus SAP acts as a natural suppressor of SLAM-mediated T cell activation, and, in the absence of SAP, T cells are constitutively activated and overproduce IFN-γ. Mutations in the SAP gene lead to abnormal T cell activation and enhanced Th1 cytokine production in mouse models and in humans: about half of patients with X-linked lympoproliferative disease (XLP) have functionally disabling SAP mutations. Acquired aplastic anemia (AA) is a bone marrow failure syndrome in which hematopoietic cell destruction is effected by cytotoxic T cells and type 1 cytokines. We have recently shown that T cells from patients with AA have increased protein levels of T-bet, resulting in IFN-γ overproduction (Solomou EE et al, Blood2006; 107:3983). IFN-γ inhibits hematopoietic stem cell proliferation and induces Fas-mediated apoptosis; stem cell depletion results in marrow hypoplasia and peripheral blood pancytopenia. We examined SAP expression as an explanation for aberrant T cell activation and extreme Th1 polarization. SAP protein expression on immunoblot was very low to absent in unstimulated T cells from 16 of 20 AA patients examined, as compared to normal levels of expression in equivalent numbers of healthy donors (p&lt;0.001). No significant differences were detected in Fyn and SLAM protein levels between AA and controls. SAP mRNA levels were also significantly decreased in T cells from those AA patients with low SAP protein levels, as determined by RT-PCR. Peripheral blood DNA samples from 18 patients with AA were analyzed for SAP mutations: three novel intronic mutations, not present in controls, were identified among 7 unrelated patients: one mutation was in the promoter region of SAP (position 106, C to T; 3 patients), and two mutations in the intron-exon junction between exons 1 and 2 (position 38975, C toT; 3 patients) and 3 and 4 (position 62771, C to A; 1 patient). IFN-γ, as measured by ELISA, in three patients with undetectable SAP protein levels was significantly increased compared to healthy controls (n=5, p&lt;0.001). Increased IFN-γ levels and Th1 polarization in AA can in part be explained by functional SAP deficiency. SAP-deficient T cells in AA would be unable to block co-stimulatory signals, leading to an activated T cell phenotype and ultimately hematopoietic cell destruction and bone marrow failure. The SAP-deficient phenotype in T cells from patients with aplastic anemia may be secondary to subtle genetic alteration in the gene’s regulation (abnormal promoter binding sites or epigenetic modulation due to mutations in introns) or as yet unidentified aberrant upstream pathways (Ets-1 and Ets-2, the transcription factors that regulate SAP expression).


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2966-2966 ◽  
Author(s):  
Manisha Bhutani ◽  
David Foureau ◽  
Tammy Cogdill ◽  
Kyle Madden ◽  
Qing Zhang ◽  
...  

Abstract BACKGROUND: Lenalidomide is an immunomodulatory drug (IMiD) with co-stimulatory effects on immune effector cells in vitro and is an approved treatment for multiple myeloma (MM), although its mode of action in patients is not well defined. We studied the impact of lenalidomide maintenance therapy, following autologous stem cell transplant (ASCT), on NK and NK-T polarization (i.e. activating or inhibitory molecules) and, T cell activation (early vs. late activation) in patients with multiple myeloma. PATIENTS AND METHODS: In this ongoing prospective study with a targeted enrollment of 28 newly diagnosed multiple myeloma patients, blood samples are being collected at 2 to 3 months post ASCT, before starting lenalidomide maintenance therapy (baseline), and serially after 1, 3 and 6 months of treatment (T+1mo, T+3mo, T+6mo). Using a 9 color flow cytometry panel, peripheral blood samples were analyzed for expression of CD3 and CD56 to define NK (CD56+ CD3-), NKT (CD56+ CD3+), and T cell (CD56- CD3+) subsets. Killer 'inhibitory' Ig-like receptors, (KiR2DS4, KiR3DL1) natural killer group 2 proteins (NKG2a, NKG2D) and natural killer p46 protein (NKp46) expression were quantified to assess polarization of NK, and NK-T cells. Programmed death receptor 1 (PD-1) and T-cell Ig and mucin receptor 3 (Tim3) expression was quantified to assess T cell activation state. Flow cytometry data were acquired on a BD FACSAria II, and analyzed using FlowJo version X software. RESULTS: Samples from 11 patients have been collected and analyzed so far (11 baseline, 6 T+1mo, 4 T+3mo). At baseline lymphoid cells represent 12-46% of white blood cells (WBC), this heterogeneity being mainly driven by a wide range of T cell relative distribution among patients (30-74 % lymphoid cells). Phenotypically, NK cells at baseline mainly express natural cytotoxicity receptors (NKp46 and NKG2D), whereas NK-T cell also express NKG2D but approximately 1/3 also express PD-1 indicating they may be functionally defective. T cells at baseline express early T cell activation markers NKG2D and approximately 1/3 also stained positive for late T cell activation marker PD-1. Lymphoid cells relative distribution among WBC tends to normalize at T+1mo of treatment (15 to 35 % of WBC) before expanding at T+3mo (35 to 43 % of WBC). Phenotypically, across the 27 immune variables analyzed, each multiple myeloma patient displayed high level of immune homeostasis after 1 or 3 months of lenalidomide treatment. Noticeably, Nkp46 expression by NK cell and PD-1 expression by NK-T cells decreased in 4/6 patients and, NKG2D expression by T cell decreased in all but one patient during lenalidomide therapy. CONCLUSION: To our knowledge, this is the first study examining the influence of lenalidomide maintenance on the comprehensive immune repertoire in the post-ASCT setting in MM patients. The wide heterogeneity of NK, NK-T and T cell distribution observed at baseline among lymphoid cells indicates the potential effect of post-ASCT immune reconstitution and immunomodulatory the impact of lenalidomide. The capacity of lenalidomide to mediate effects on several immune cells raises the question as to which, if any, of these changes correlate with clinical responses. In our study, serially collected data from each patient, when completed would determine the impact of lenalidomide immunomodulatory effect of therapeutic efficacy and PFS duration in relation to immune reconstitution stage. Disclosures Cogdill: Millennium: Speakers Bureau; Onyx: Speakers Bureau; Celgene: Speakers Bureau; Novartis: Speakers Bureau. Ghosh:Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Usmani:Sanofi: Honoraria, Research Funding; Millennium: Honoraria, Speakers Bureau; Onyx: Honoraria, Research Funding, Speakers Bureau; Pharmacyclics: Research Funding; Celgene: Honoraria, Speakers Bureau; Janssen Oncology: Honoraria, Research Funding; Array BioPharma: Honoraria, Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2154-2154
Author(s):  
Jing-Xia Li ◽  
Jian-Ming Li ◽  
Edmund K Waller

Abstract Introduction: Pre-clinical murine experiments and clinical data from allogeneic bone marrow transplantation (allo-BMT) have shown that increased numbers of plasmacytoid dendritic cells (pDC) in the bone marrow graft results in better clinical outcomes with less severe graft-versus-host disease (GvHD) and improved survival. The mechanism by which donor pDC modulate GvHD is unknown. Knowing that vasoactive intestinal polypeptide (VIP) is an immunosuppressive peptide , we reasoned that VIP signaling might play a role in regulating T-cell activation and expansion, and the VIP pathway may be a potential therapeutic target for regulating GvHD in allo-BMT. We have tested the hypothesis that VIP synthesis by donor pDCs can modulate T cell alloreactivity. Methods: To explore the mechanisms by which pDC and VIP signaling regulate T cell activation in murine allo-BMT, we prepared B6-background donor cell grafts and transplanted them into lethally irradiated B10.BR recipients. In experiment 1, recipients were transplanted with grafts containing the combination of 5 x 103 VIP-GFP hematopoietic stem cells (HSC) and 3 x 106 VIP-wild type (VIP-WT) or VIP-knock out (VIP-KO) splenocytes. At day 7, splenocytes were isolated for flow cytometric analysis looking for GFP signal, which represents VIP-promotor activity. Experiment 2 used combinations of 5 x 103 VIP-WT HSC, 1 x 106 luciferase+ T cells, and 50 x 103 VIP-WT or VIP-KO pDC from B6 as donor grafts. Recipients were monitored for survival and GvHD based on fur texture, posture, activity, skin integrity and weight loss. T cell expansion was measured by bioluminescent imaging (BLI). Serum cytokines from bleeds at day 3 and day 8 post-transplant were analyzed using a Luminex 38 plex panel. Some recipients were euthanized on day 3 for intracellular cytokine analysis of splenic T cells. Results: In experiment 1, 7 days post-transplant, analysis of splenocytes from all mice showed increased activity of the VIP gene promoter in donor pDC that were derived from HSC, compared to other cell types. The VIP promoter signal was also stronger in donor HSC-derived pDCs, if originally transplanted with VIP-KO splenocytes. In experiment 2 over 70% of mice receiving HSC+T+VIP-WT pDC in the BM graft survived to day 100 post-transplant, while those getting VIP-KO pDC instead only had 30% survival (Fig 1A). All surviving recipients were fully engrafted by day 30. BLI revealed greater total T-cell proliferation (measured as radiance) in recipients of VIP-KO pDC (Fig 1B). Furthermore, recipients of VIP-KO pDC had more severe acute GvHD, with increased weight loss and GvHD clinical scores (Fig 1C, 1D). Some recipients were euthanized and their serum were collected for cytokine analysis on day 8 post-transplant, which showed up-regulation of pro-inflammatory or chemotactic cytokines MCP1, IL-1, IL-12, IL-17 in T cells co-transplanted with VIP-KO pDC compared to WT pDC. Conclusion: The present findings show that: 1) VIP is produced by donor pDC early after allo-BMT; 2) absence of VIP production by donor pDC leads to increased T-cell expansion in a murine allo-BMT model. Thus the pDC-T cell VIP signaling pathway is a critical element in controlling donor T cell alloreactivity after allo-BMT. Future studies will include VIP qPCR to confirm VIP production in donor pDC post-transplant, and determine the mechanism by which VIP production by pDC regulates T cell activity and modulates GvHD. Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.


2002 ◽  
Vol 195 (7) ◽  
pp. 811-823 ◽  
Author(s):  
Dietrich Conze ◽  
Troy Krahl ◽  
Norman Kennedy ◽  
Linda Weiss ◽  
Joanne Lumsden ◽  
...  

The c-Jun NH2-terminal kinase (JNK) signaling pathway is induced by cytokines and stress stimuli and is implicated in cell death and differentiation, but the specific function of this pathway depends on the cell type. Here we examined the role of JNK1 and JNK2 in CD8+ T cells. Unlike CD4+ T cells, the absence of JNK2 causes increased interleukin (IL)-2 production and proliferation of CD8+ T cells. In contrast, JNK1-deficient CD8+ T cells are unable to undergo antigen-stimulated expansion in vitro, even in the presence of exogenous IL-2. The hypoproliferation of these cells is associated with impaired IL-2 receptor α chain (CD25) gene and cell surface expression. The reduced level of nuclear activating protein 1 (AP-1) complexes in activated JNK1-deficient CD8+ T cells can account for the impaired IL-2 receptor α chain gene expression. Thus, JNK1 and JNK2 play different roles during CD8+ T cell activation and these roles differ from those in CD4+ T cells.


Sign in / Sign up

Export Citation Format

Share Document