Anti-CD3 Fab Fragments Enhance Tumor Killing by Human γδ T Cells Independent of Nck Recruitment to the γδ T Cell Antigen Receptor

Claudia Juraske, Piyamaporn Wipa, Anna Morath, Jose Villacorta Hidalgo, Frederike A Hartl, Katrin Raute, Hans-Heinrich Oberg, Daniela Wesch, Paul Fisch, Susana Minguet, Sutatip Pongcharoen, Wolfgang W Schamel, Claudia Juraske, Piyamaporn Wipa, Anna Morath, Jose Villacorta Hidalgo, Frederike A Hartl, Katrin Raute, Hans-Heinrich Oberg, Daniela Wesch, Paul Fisch, Susana Minguet, Sutatip Pongcharoen, Wolfgang W Schamel

Abstract

T lymphocytes expressing the γδ T cell receptor (γδ TCR) can recognize antigens expressed by tumor cells and subsequently kill these cells. γδ T cells are indeed used in cancer immunotherapy clinical trials. The anti-CD3ε antibody UCHT1 enhanced the in vitro tumor killing activity of human γδ T cells by an unknown molecular mechanism. Here, we demonstrate that Fab fragments of UCHT1, which only bind monovalently to the γδ TCR, also enhanced tumor killing by expanded human Vγ9Vδ2 γδ T cells or pan-γδ T cells of the peripheral blood. The Fab fragments induced Nck recruitment to the γδ TCR, suggesting that they stabilized the γδ TCR in an active CD3ε conformation. However, blocking the Nck-CD3ε interaction in γδ T cells using the small molecule inhibitor AX-024 neither reduced the γδ T cells' natural nor the Fab-enhanced tumor killing activity. Likewise, Nck recruitment to CD3ε was not required for intracellular signaling, CD69 and CD25 up-regulation, or cytokine secretion by γδ T cells. Thus, the Nck-CD3ε interaction seems to be dispensable in γδ T cells.

Keywords: AX-024; Fab fragments; Nck; T cell antigen receptor; activation; signaling; tumor; γδ T cells.

Figures

Figure 1
Figure 1
UCHT1 increased tumor killing by γδ T cells. 51Cr-labeled Daudi and Raji cells were incubated with zoledronate-expanded human γδ T cells without (−) or with the addition of 5 µg/ml UCHT1 using an effector to target (T cell to tumor cell) ratio of 12.5:1. After 5 h of co-culture, the amount of released 51Cr was measured in the supernatant by γ-ray spectroscopy. Data represent mean ± SD of triplicate wells (n = 3). Significance was determined by unpaired t-test, two-tailed between untreated γδ T cells and treated samples.
Figure 2
Figure 2
Using its SH3.1(Nck) domain Nck binds to the UCHT1-stimulated γδ T cell antigen receptor (TCR). (A) Schematic of the SH3.1(Nck) pull down assay. (B) Zoledronate-expanded γδ T cells were left untreated (−) or stimulated with 5 µg/ml UCHT1 at 37°C for 5 min. The cellular lysates were incubated with SH3.1(Nck)-coupled beads and bound proteins separated by SDS-PAGE along with aliquots of the cellular lysate. Western blotting was done using anti-CD3ζ and anti-GST antibodies. (C) The normalized ratio of the band intensities of CD3ζ to GST in the SH3.1(Nck) pull down is plotted. Data of three independent experiments were used to calculate the mean ± SD; statistics was done by two-tailed t-test. (D) Schematic of the in situ proximity ligation assay (PLA) using anti-CD3ε and anti-Nck antibodies. (E) Close proximity between the TCR and Nck was detected by in situ PLA. Zoledronate-expanded γδ T cells were either left untreated (−) or treated with 5 µg/ml UCHT1 in the absence or presence of 10 nM AX-024 at 37°C for 5 min. After fixation and permeabilization, PLA was performed using the primary antibodies goat anti-CD3ε (M20) and rabbit anti-Nck1, and the corresponding secondary antibodies. Nuclei were stained with DAPI. (F) The corresponding quantification of the red PLA dots and the mean ± SEM is displayed; statistics was done by two-tailed t-test. For each condition 500 cells were analyzed. Three independent experiments were performed (n = 3).
Figure 3
Figure 3
Preparation of UCHT1 Fab fragments. (A) The anti-CD3ε antibody UCHT1 was digested with papain using a kit from Thermo Fisher Scientific. The Fab fragments were purified with protein A coupled beads. The complete antibody (lane 1), the first and second digestions (lanes 2 and 3), and the purified Fab fragment (lane 4) were separated by reducing SDS-PAGE followed by Coomassie staining (n > 3). (B) Jurkat Vγ9Vδ2 cells were labeled with Fluo-3 AA and Fura Red AM and the baseline of cellular Ca2+ was measured by flow cytometry for 1 min. The indicated reagents were then added and Ca2+ levels were measured for additional 6 min (black lines). The gray lines represent baseline Ca2+ level without addition of any stimuli (n > 3). (C) Concanavalin A expanded γδ T cells were stained with UCHT1, Fab, Fabred, or left untreated, followed by an APC-labeled anti-mouse IgG antibody as a secondary reagent. Fluorescence intensities were quantified by flow cytometry (n > 3).
Figure 4
Figure 4
Fab and Fabred fragments enhanced tumor killing by γδ T cells. 51Cr-labeled Daudi and Raji cells were incubated with zoledronate (A) or concanavalin A (B) expanded γδ T cells in triplicates without (−) or with 5 µg/ml UCHT1, 3.33 µg/ml Fab, or 3.33 µg/ml Fabred (these concentrations were chosen to obtain equimolar amounts of the different reagents). The effector to target ratio was 12.5:1. After 5 h of co-culture, the amount of released 51Cr was measured in the supernatant by γ-ray spectroscopy (n = 3). (C) Concanavalin A expanded γδ T cells from four different donors (HD1, 2, 3, and 4) were used in a chromium-release assay using Daudi cells as target as in (A) (n = 1). Data represent mean ± SD of triplicate wells. Significance was determined by unpaired t-test, two-tailed between untreated γδ T cells and UCHT1, Fab or Fabred-treated samples.
Figure 5
Figure 5
UCHT1 Fab induced the recruitment of Nck to the γδ T cell antigen receptor. (A) The proximity ligation assay as in Figure 2 was performed using 5 µg/ml UCHT1, 3.33 µg/ml Fab, and 3.33 µg/ml Fabred in the absence or presence of 10 nM AX-024. (B) The data of (A) were analyzed as in Figure 2F; 700 cells were analyzed per condition. Two independent experiments were performed (n = 2).
Figure 6
Figure 6
Tumor killing by γδ T cells is independent of Nck recruitment to the γδ T cell antigen receptor. 51Cr-labeled Daudi cells were incubated with concanavalin A (A) or zoledronate (B) expanded γδ T cells without (−) or with 5 µg/ml UCHT1, 3.33 µg/ml Fab, or 3.33 µg/ml Fabred. In addition, 10 or 100 nM of AX-024 was included or not. The effector to target ratio was 12.5:1. After 5 h of co-culture, the amount of released 51Cr was measured in the supernatant by γ-ray spectroscopy. Data represent mean ± SD of triplicates (n = 3). Significance was determined by unpaired t-test, two-tailed between untreated samples and samples treated with AX-024.
Figure 7
Figure 7
The up-regulation of activation markers and cytokines is independent of Nck recruitment to the γδ T cell antigen receptor (TCR). Daudi and Raji cells were incubated with zoledronate-expanded γδ T cells without (−) or with 5 µg/ml UCHT1, 3.33 µg/ml Fab, or 3.33 µg/ml Fabred. In addition, 10 nM AX-024 was included or not. The γδ T cell to target cell ratio was 12.5:1. The cells were co-cultured at 37°C and 5% CO2 for 18 h. Subsequently, cells were either stained with anti-CD25 and anti-γ/δTCR (A,B) or with anti-CD69 and anti-γ/δTCR (C) antibodies. Fluorescence intensities were quantified by flow cytometry (A) and the mean fluorescence intensity is displayed (B,C). Data represent mean ± SD of triplicates (n = 3). Significance was determined by unpaired t-test, two-tailed between untreated samples and samples treated with AX-024. (D,E) The experiments were performed as in (A). After co-culturing of the cells for 18 h, the supernatants were harvested. TNFα (D) and IFNγ (E) concentrations were quantified by enzyme linked immunosorbent assay.
Figure 8
Figure 8
Calcium signaling is enhanced by Fab fragments and does not require the Nck-γδ T cell antigen receptor (TCR) interaction. (A) G8 γδ TCR-expressing Jurkat cells were stained with the γδ TCR-specific antibody GL3 and analyzed by flow cytometry. As controls, the GL3-stained parental Jurkat cells and unstained G8 γδ TCR-expressing cells are shown (n > 3). (B) Jurkat cells expressing the G8 γδ TCR were stimulated with the G8 ligand T22 tetramers (T22t), UCHT1 Fab, or a combination of both T22t and Fab. Intracellular calcium was measured using the dyes Fluo-3 and Fura-red by flow cytometry. Calcium influx is depicted as the median ratio of Fluo-3 to Fura-red fluorescence over time. As a negative control, steptavidin was added to the cells. The arrow indicates addition of the stimuli (n = 3). (C) The experiment was performed as in (C) with the difference that cells without and with 10 nM AX-024 were stimulated with the combination of T22t and Fab. As a control, PBS was added to the cells (n = 3).

References

    1. Groh V, Steinle A, Bauer S, Spies T. Recognition of stress-induced MHC molecules by intestinal epithelial γδ T cells. Science (1998) 279:1737–40.10.1126/science.279.5357.1737
    1. Uldrich AP, Le Nours J, Pellicci DG, Gherardin NA, McPherson KG, Lim RT, et al. CD1d-lipid antigen recognition by the γδ TCR. Nat Immunol (2013) 14:1137–45.10.1038/ni.2713
    1. Vantourout P, Hayday A. Six-of-the-best: unique contributions of γδ T cells to immunology. Nat Rev Immunol (2013) 13:88–100.10.1038/nri3384
    1. Chien Y, Konigshofer Y. Antigen recognition by γδ T cells. Immunol Rev (2007) 215:46–58.10.1111/j.1600-065X.2006.00470.x
    1. Bonneville M, O’Brien RL, Born WK. γδ T cell effector functions: a blend of innate programming and acquired plasticity. Nat Rev Immunol (2010) 10:467–78.10.1038/nri2781
    1. Zou C, Zhao P, Xiao Z, Han X, Fu F, Fu L. γδ T cells in cancer immunotherapy. Oncotarget (2017) 8:8900–9.10.18632/oncotarget.13051
    1. Chitadze G, Oberg HH, Wesch D, Kabelitz D. The ambiguous role of γδ T lymphocytes in antitumor immunity. Trends Immunol (2017) 38:668–78.10.1016/j.it.2017.06.004
    1. Bukowski JF, Morita CT, Band H, Brenner MB. Crucial role of TCR γ chain junctional region in prenyl pyrophosphate antigen recognition by γδ T cells. J Immunol (1998) 161:286–93.
    1. Bukowski JF, Morita CT, Tanaka Y, Bloom BR, Brenner MB, Band H. Vγ2Vδ2 TCR-dependent recognition of non-peptide antigens and Daudi cells analyzed by TCR gene transfer. J Immunol (1995) 154:998–1006.
    1. Constant P, Davodeau F, Peyrat MA, Poquet Y, Puzo G, Bonneville M, et al. Stimulation of human γδ T cells by nonpeptidic mycobacterial ligands. Science (1994) 264:267–70.10.1126/science.8146660
    1. Tanaka Y, Morita CT, Tanaka Y, Nieves E, Brenner MB, Bloom BR. Natural and synthetic non-peptide antigens recognized by human γδ T cells. Nature (1995) 375:155–8.10.1038/375155a0
    1. Espinosa E, Belmant C, Pont F, Luciani B, Poupot R, Romagne F, et al. Chemical synthesis and biological activity of bromohydrin pyrophosphate, a potent stimulator of human γδ T cells. J Biol Chem (2001) 276:18337–44.10.1074/jbc.M100495200
    1. Fisch P, Meuer E, Pende D, Rothenfusser S, Viale O, Kock S, et al. Control of B cell lymphoma recognition via natural killer inhibitory receptors implies a role for human Vγ9/Vδ2 T cells in tumor immunity. Eur J Immunol (1997) 27:3368–79.10.1002/eji.1830271236
    1. Gober HJ, Kistowska M, Angman L, Jenö P, Mori L, De Libero G. Human T cell receptor γδ cells recognize endogenous mevalonate metabolites in tumor cells. J Exp Med (2003) 197:163–8.10.1084/jem.20021500
    1. Silva-Santos B, Serre K, Norell H. γδ T cells in cancer. Nat Rev Immunol (2015) 15:683–91.10.1038/nri3904
    1. Roelofs AJ, Jauhiainen M, Monkkonen H, Rogers MJ, Monkkonen J, Thompson K. Peripheral blood monocytes are responsible for γδ T cell activation induced by zoledronic acid through accumulation of IPP/DMAPP. Br J Haematol (2009) 144:245–50.10.1111/j.1365-2141.2008.07435.x
    1. Benzaid I, Monkkonen H, Bonnelye E, Monkkonen J, Clezardin P. In vivo phosphoantigen levels in bisphosphonate-treated human breast tumors trigger Vgamma9Vdelta2 T-cell antitumor cytotoxicity through ICAM-1 engagement. Clin Cancer Res (2012) 18:6249–59.10.1158/1078-0432.CCR-12-0918
    1. Kabelitz D, Wesch D, Pitters E, Zoller M. Characterization of tumor reactivity of human Vγ9Vδ2 γδ T cells in vitro and in SCID mice in vivo. J Immunol (2004) 173:6767–76.10.4049/jimmunol.173.11.6767
    1. Dieli F, Vermijlen D, Fulfaro F, Caccamo N, Meraviglia S, Cicero G, et al. Targeting human γδ T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Res (2007) 67:7450–7.10.1158/0008-5472.CAN-07-0199
    1. Meraviglia S, Eberl M, Vermijlen D, Todaro M, Buccheri S, Cicero G, et al. In vivo manipulation of Vγ9Vδ2 T cells with zoledronate and low-dose IL-2 for immunotherapy of advanced breast cancer patients. Clin Exp Immunol (2010) 161:290–7.10.1111/j.1365-2249.2010.04167.x
    1. Oberg HH, Grage-Griebenow E, dam-Klages SA, Jerg E, Peipp M, Kellner C, et al. Monitoring and functional characterization of the lymphocytic compartment in pancreatic ductal adenocarcinoma patients. Pancreatology (2016) 16:1069–79.10.1016/j.pan.2016.07.008
    1. Oberg HH, Kellner C, Gonnermann D, Peipp M, Peters C, Sebens S, et al. γδ T cell activation by bispecific antibodies. Cell Immunol (2015) 296:41–9.10.1016/j.cellimm.2015.04.009
    1. Oberg HH, Peipp M, Kellner C, Sebens S, Krause S, Petrick D, et al. Novel bispecific antibodies increase γδ T-cell cytotoxicity against pancreatic cancer cells. Cancer Res (2014) 74:1349–60.10.1158/0008-5472.CAN-13-0675
    1. Vavassori S, Kumar A, Wan GS, Ramanjaneyulu GS, Cavallari M, El Daker S, et al. Butyrophilin 3A1 binds phosphorylated antigens and stimulates human γδ T cells. Nat Immunol (2013) 14:908–16.10.1038/ni.2665
    1. Sandstrom A, Peigne CM, Leger A, Crooks JE, Konczak F, Gesnel MC, et al. The intracellular B30.2 domain of butyrophilin 3A1 binds phosphoantigens to mediate activation of human Vγ9Vδ2 T cells. Immunity (2014) 40:490–500.10.1016/j.immuni.2014.03.003
    1. Gil D, Schamel WW, Montoya M, Sanchez-Madrid F, Alarcon B. Recruitment of Nck by CD3 epsilon reveals a ligand-induced conformational change essential for T cell receptor signaling and synapse formation. Cell (2002) 109:901–12.10.1016/S0092-8674(02)00799-7
    1. Swamy M, Beck-Garcia K, Beck-Garcia E, Hartl FA, Morath A, Yousefi OS, et al. A cholesterol-based allostery model of T cell receptor phosphorylation. Immunity (2016) 44:1091–101.10.1016/j.immuni.2016.04.011
    1. Schamel WW, Alarcon B, Hofer T, Minguet S. The allostery model of TCR regulation. J Immunol (2017) 198:47–52.10.4049/jimmunol.1601661
    1. Risueno RM, Gil D, Fernandez E, Sanchez-Madrid F, Alarcon B. Ligand-induced conformational change in the T-cell receptor associated with productive immune synapses. Blood (2005) 106:601–8.10.1182/blood-2004-12-4763
    1. Minguet S, Swamy M, Alarcon B, Luescher IF, Schamel WW. Full activation of the T cell receptor requires both clustering and conformational changes at CD3. Immunity (2007) 26:43–54.10.1016/j.immuni.2006.10.019
    1. Martínez-Martín N, Risueño RM, Morreale A, Zaldívar I, Fernández-Arenas E, Herranz F, et al. Cooperativity between T cell receptor complexes revealed by conformational mutants of CD3epsilon. Sci Signal (2009) 2:ra43.10.1126/scisignal.2000402
    1. Blanco R, Borroto A, Schamel WW, Pereira P, Alarcon B. Conformational changes in the T cell receptor differentially determine T cell subset development in mice. Sci Signal (2014) 7:ra115.10.1126/scisignal.2005650
    1. Paensuwan P, Hartl FA, Yousefi OS, Ngoenkam J, Wipa P, Beck-Garcia E, et al. Nck binds to the T cell antigen receptor using its SH3.1 and SH2 domains in a cooperative manner, promoting TCR functioning. J Immunol (2016) 196:448–58.10.4049/jimmunol.1500958
    1. Borroto A, Arellano I, Dopfer EP, Prouza M, Suchanek M, Fuentes M, et al. Nck recruitment to the TCR required for ZAP70 activation during thymic development. J Immunol (2013) 190:1103–12.10.4049/jimmunol.1202055
    1. Borroto A, Arellano I, Blanco R, Fuentes M, Orfao A, Dopfer EP, et al. Relevance of Nck-CD3epsilon interaction for T cell activation in vivo. J Immunol (2014) 192:2042–53.10.4049/jimmunol.1203414
    1. Borroto A, Reyes-Garau D, Jimenez MA, Carrasco E, Moreno B, Martinez-Pasamar S, et al. First-in-class inhibitor of the T cell receptor for the treatment of autoimmune diseases. Sci Transl Med (2016) 8:370ra184.10.1126/scitranslmed.aaf2140
    1. Minguet S, Schamel WWA. A permissive geometry model for TCR-CD3 activation. Trends Biochem Sci (2008) 33:51–7.10.1016/j.tibs.2007.10.008
    1. Chang TW, Kung PC, Gingras SP, Goldstein G. Does OKT3 monoclonal antibody react with an antigen-recognition structure on human T cells? Proc Natl Acad Sci U S A (1981) 78:1805–8.10.1073/pnas.78.3.1805
    1. Kaye J, Janeway CA, Jr. The Fab fragment of a directly activating monoclonal antibody that precipitates a disulfide-linked heterodimer from a helper T cell clone blocks activation by either allogeneic Ia or antigen and self-Ia. J Exp Med (1984) 159:1397–412.10.1084/jem.159.5.1397
    1. Cordoba SP, Choudhuri K, Zhang H, Bridge M, Basat AB, Dustin ML, et al. The large ectodomains of CD45 and CD148 regulate their segregation from and inhibition of ligated T-cell receptor. Blood (2013) 121:4295–302.10.1182/blood-2012-07-442251
    1. Dopfer EP, Hartl FA, Oberg HH, Siegers GM, Yousefi OS, Kock S, et al. The CD3 conformational change in the γδ T cell receptor is not triggered by antigens but can be enforced to enhance tumor killing. Cell Rep (2014) 7:1704–15.10.1016/j.celrep.2014.04.049
    1. Soderberg O, Gullberg M, Jarvius M, Ridderstrale K, Leuchowius KJ, Jarvius J, et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat Methods (2006) 3:995–1000.10.1038/nmeth947
    1. Alibaud L, Arnaud J, Llobera R, Rubin B. On the role of CD3δ chains in TCRγδ/CD3 complexes during assembly and membrane expression. Scand J Immunol (2001) 54:155–62.10.1046/j.1365-3083.2001.00938.x
    1. Siegers GM, Swamy M, Fernandez-Malave E, Minguet S, Rathmann S, Guardo AC, et al. Different composition of the human and the mouse γδ T cell receptor explains different phenotypes of CD3γ and CD3δ-immunodeficiencies. J Exp Med (2007) 204:2537–44.10.1084/jem.20070782102207c
    1. Siegers GM, Ribot EJ, Keating A, Foster PJ. Extensive expansion of primary human γδ T cells generates cytotoxic effector memory cells that can be labeled with Feraheme for cellular MRI. Cancer Immunol Immunother (2012) 62:571–83.10.1007/s00262-012-1353-y
    1. Wesch D, Marx S, Kabelitz D. Comparative analysis of alpha beta and γδ T cell activation by Mycobacterium tuberculosis and isopentenyl pyrophosphate. Eur J Immunol (1997) 27:952–6.10.1002/eji.1830270422
    1. Kondo M, Sakuta K, Noguchi A, Ariyoshi N, Sato K, Sato S, et al. Zoledronate facilitates large-scale ex vivo expansion of functional γδ T cells from cancer patients for use in adoptive immunotherapy. Cytotherapy (2008) 10:842–56.10.1080/14653240802419328
    1. Crowley MP, Fahrer AM, Baumgarth N, Hampl J, Gutgemann I, Teyton L, et al. A population of murine γδ T cells that recognize an inducible MHC class Ib molecule. Science (2000) 287:314–6.10.1126/science.287.5451.314
    1. Adams EJ, Chien YH, Garcia KC. Structure of a γδ T cell receptor in complex with the nonclassical MHC T22. Science (2005) 308:227–31.10.1126/science.1106885
    1. Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med (2015) 21:938–45.10.1038/nm.3909
    1. Deniger DC, Moyes JS, Cooper LJ. Clinical applications of γδ T cells with multivalent immunity. Front Immunol (2014) 5:636.10.3389/fimmu.2014.00636
    1. Fournie JJ, Sicard H, Poupot M, Bezombes C, Blanc A, Romagne F, et al. What lessons can be learned from γδ T cell-based cancer immunotherapy trials? Cell Mol Immunol (2013) 10:35–41.10.1038/cmi.2012.39
    1. Paul S, Lal G. Regulatory and effector functions of γδ T cells and their therapeutic potential in adoptive cellular therapy for cancer. Int J Cancer (2016) 139:976–85.10.1002/ijc.30109
    1. Courtney AH, Lo WL, Weiss A. TCR signaling: mechanisms of initiation and propagation. Trends Biochem Sci (2018) 43:108–23.10.1016/j.tibs.2017.11.008
    1. Lafont V, Liautard J, Sable-Techene M, Sainte-Marie Y, Favero J. Isopentenyl pyrophosphate, a mycobacterial non-peptidic antigen, triggers delayed and highly sustained signaling in human γδ T lymphocytes without inducing down-modulation of T cell antigen receptor. J Biol Chem (2001) 276:15961–7.10.1074/jbc.M008684200
    1. Correia DV, d’Orey F, Cardoso BA, Lanca T, Grosso AR, deBarros A, et al. Highly active microbial phosphoantigen induces rapid yet sustained MEK/Erk- and PI-3K/Akt-mediated signal transduction in anti-tumor human γδ T-cells. PLoS One (2009) 4:e5657.10.1371/journal.pone.0005657
    1. Nedellec S, Sabourin C, Bonneville M, Scotet E. NKG2D costimulates human V gamma 9V delta 2 T cell antitumor cytotoxicity through protein kinase C theta-dependent modulation of early TCR-induced calcium and transduction signals. J Immunol (2010) 185:55–63.10.4049/jimmunol.1000373
    1. Thedrez A, Sabourin C, Gertner J, Devilder MC, Allain-Maillet S, Fournie JJ, et al. Self/non-self discrimination by human γδ T cells: simple solutions for a complex issue? Immunol Rev (2007) 215:123–35.10.1111/j.1600-065X.2006.00468.x
    1. Cipriani B, Knowles H, Chen L, Battistini L, Brosnan CF. Involvement of classical and novel protein kinase C isoforms in the response of human Vγ9Vδ2 T cells to phosphate antigens. J Immunol (2002) 169:5761–70.10.4049/jimmunol.169.10.5761

Source: PubMed

3
Abonnieren