Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance

Douglas C Palmer, Geoffrey C Guittard, Zulmarie Franco, Joseph G Crompton, Robert L Eil, Shashank J Patel, Yun Ji, Nicholas Van Panhuys, Christopher A Klebanoff, Madhusudhanan Sukumar, David Clever, Anna Chichura, Rahul Roychoudhuri, Rajat Varma, Ena Wang, Luca Gattinoni, Francesco M Marincola, Lakshmi Balagopalan, Lawrence E Samelson, Nicholas P Restifo, Douglas C Palmer, Geoffrey C Guittard, Zulmarie Franco, Joseph G Crompton, Robert L Eil, Shashank J Patel, Yun Ji, Nicholas Van Panhuys, Christopher A Klebanoff, Madhusudhanan Sukumar, David Clever, Anna Chichura, Rahul Roychoudhuri, Rajat Varma, Ena Wang, Luca Gattinoni, Francesco M Marincola, Lakshmi Balagopalan, Lawrence E Samelson, Nicholas P Restifo

Abstract

Improving the functional avidity of effector T cells is critical in overcoming inhibitory factors within the tumor microenvironment and eliciting tumor regression. We have found that Cish, a member of the suppressor of cytokine signaling (SOCS) family, is induced by TCR stimulation in CD8(+) T cells and inhibits their functional avidity against tumors. Genetic deletion of Cish in CD8(+) T cells enhances their expansion, functional avidity, and cytokine polyfunctionality, resulting in pronounced and durable regression of established tumors. Although Cish is commonly thought to block STAT5 activation, we found that the primary molecular basis of Cish suppression is through inhibition of TCR signaling. Cish physically interacts with the TCR intermediate PLC-γ1, targeting it for proteasomal degradation after TCR stimulation. These findings establish a novel targetable interaction that regulates the functional avidity of tumor-specific CD8(+) T cells and can be manipulated to improve adoptive cancer immunotherapy.

Figures

Figure 1.
Figure 1.
Cish induction is TCR stimulation dependent. (A) Induction of Cish gene expression using real-time PCR at indicated times after αCD3 stimulation of naive CD8+ T cells. Results shown as means ± SEM n = 3; two independent experiments. (B) Western Blot of Cish protein expression after αCD3 stimulation of naive CD8+ T cells at indicated times; three independent experiments. (C) Relative Cish mRNA expression in different CD8+ T cell subsets using real-time PCR; Naive (Tn; CD62L+CD44−), in vivo–derived central memory (Tcm; CD62L+CD44+), and effector memory (Tem; CD62L−CD44+) after vaccination. n = 3; three independent experiments. (D) Representative FACS blot of tumor-resident pmel-1 thy1.1+ CD8+ T cells 7 d after ACT. n = 3; two independent experiments. (E and F) Representative FACS blot of Cish expression by intracellular staining of tissue-resident pmel-1 CD8+ T cells in antigen-negative nondraining axillary lymph node (Irr LN) or antigen-positive tumor 7 d after ACT. *, P < 0.05 by unpaired Student’s t test. n = 3; two independent experiments. (G) Schematic of Cish knockout targeting construct (B, BamHI; E, EcoRI; H, HindIII; P, PstI; X, XhoI). (H) PCR confirmation of genotype. Gel electrophoresis of DNA products after PCR amplification. n = 300+. (I) Representative FACS blot of CD8+ and CD4+ thymocytes from Cish−/− or WT mice, enumerated in J. Values represent mean ± SEM. P > 0.05 by unpaired Student’s t test. n = 3; three independent experiments. (K) Enumeration and flow cytometric evaluation of CD4+ and CD8+ splenocytes for each genotype. Values represent mean ± SEM. P > 0.05 by unpaired Student’s t test. n = 3; three independent experiments.
Figure 2.
Figure 2.
Cish inhibits CD8+ T cell expansion and functional avidity. (A) Phenotypic memory analysis of CD8+ T cells from Cish−/− and WT pmel-1 littermate mice using flow cytometry. Values represent mean ± SEM. P > 0.05 by unpaired Student’s t test. n = 3; three independent experiments. (B) Enumeration of T cell expansion 6 d after stimulation with hgp10025-33 peptide-pulsed splenocytes. Values represent mean ± SEM. **, P < 0.01 by unpaired Student’s t test. n = 3; three independent experiments. (C) Evaluation of apoptosis of primed Cish−/− or WT CD8+ T cells as assessed by flow cytometric evaluation of 7-AAD and Annexin V staining after αCD3 restimulation at times indicated. n = 3; three independent experiments. (D–F) Enzyme-linked immunosorbent assay (ELISA) evaluation of IFN-γ, TNF or IL-2 in supernatants after an overnight co-culture of primed Cish−/− or WT pmel-1 CD8+ T cells with peptide-pulsed splenocytes. Values are shown as means ± SEM. ****, P < 0.0001 by two-way ANOVA. n = 3; three independent experiments. (G) Representative FACS blot of intracellular IFN-γ, TNF, and IL-2 in Cish−/− or WT pmel-1 CD8+ T cells 6 h after αCD3 stimulation. n = 3; three independent experiments. (H) Assessment of cytokine polyfunctionality in Cish−/− or WT pmel-1 CD8+ T cells from (G) using Boolean gating strategy. Data are shown as total percentage of two or three concomitant cytokines present after antigenic stimulation. n = 3; three independent experiments.
Figure 3.
Figure 3.
Cish deletion augments CD8+ T cell–mediated tumor immunity. (A–E) 7 d after s.c. implantation of 5 × 105 B16 melanoma cells, C57BL/6 tumor-bearing mice received the adoptive transfer (ACT) of 106 Cish−/−, WT pmel-1 naive CD8+ T cells, or no cells (NT) in conjunction with rhgp100 VV (107 pfu) and IL-2 (2 × 106 IU BID for 3 d). Tumor progression, survival, and expansion of pmel-1 T cells were followed. (A) Tumor growth as assessed by measurement of the perpendicular diameters over time. Values are shown as means ± SEM. ****, P < 0.0001 by two-way ANOVA. n = 5 per group; 10 independent experiments. (B) Kaplan-Meier survival curve after ACT in A. **, P < 0.01 by Log-rank test for trend. n = 5–6 per group; 10 independent experiments. (C) Induction of ocular autoimmunity 6 d after ACT as assessed by combinatorial scoring of severity of iridiocyclitis, vitritis, and choroiditis as described in Materials and methods. ****, P < 0.0001. n = 6; two independent experiments. (D) In vivo growth kinetics of congenically marked T cells after ACT into C57BL/6 recipients as assessed by flow cytometry in conditions identical to (A). Values are shown as means ± SEM. ****, P < 0.0001 by two-way ANOVA. n = 3; three independent experiments. (E) Ex vivo Annexin V staining by flow cytometry of congenically marked Cish−/− or WT pmel-1 T cells from the spleen 6 d after ACT. *, P < 0.05 by unpaired Student’s t test. n = 3; two independent experiments.
Figure 4.
Figure 4.
Cishdeletion augments long-term CD8+ T cell–intrinsic tumor killing. (A–D) Growth of B16 melanoma in Rag1−/− tumor-bearing hosts after ACT of 2.5 × 105 indicated pmel-1 CD8+ T cells in conjunction with rhgp100 VV (3 × 106 pfu) and IL-2 (2 × 105 IU BID for 3 d) or no cell transfer (NT). (A) Tumor growth as assessed by measurement of the perpendicular diameters over time. Values are shown as means ± SEM. ****, P < 0.0001 by two-way ANOVA. n = 5 per group; three independent experiments. (B) Outgrowth of amelanotic tumors from A in five out of five mice, 50+ d after ACT; two independent experiments. (C) IFN-γ production assessed by ELISA 7 d after ACT, after co-culture with ex vivo enriched congenically marked Cish−/− or WT pmel-1 T cells with cognate peptide-pulsed splenocytes. T cells were normalized for cell number. Values are shown as means ± SEM. ****, P < 0.0001 by two-way ANOVA. n = 3; two independent experiments. (D) Growth of B16 melanoma in Rag1−/− tumor-bearing hosts as assessed by measurement of the perpendicular diameters over time after treatment. ACT of 2.5 × 105Cish−/− pmel-1 CD8+ T cells or no cell transfer (NT) with or without administration of αCD8-depleting antibody 38 d after transfer. Values are shown as means ± SEM. **, P < 0.01 by two-way ANOVA. n = 5; two independent experiments.
Figure 5.
Figure 5.
Knockdown of Cish in mice and man confers enhanced CD8+ T cell tumor reactivity. (A) IFN-γ evaluation by ELISA after an overnight co-culture of Cish shmiR or control shmiR retroviral transduced CD8+ pmel-1 WT T cells. Values are shown as means ± SEM. ****, P < 0.0001 by two-way ANOVA. n = 3; two independent experiments. (B) Growth of B16 melanoma in C57BL/6 tumor-bearing hosts as assessed by measurement of the perpendicular diameters over time after treatment. ACT of 1 × 106 of retroviral transduced Cish shmiR WT, control shmiR WT, or control Cish−/− pmel-1 T cells in conjunction with rhgp100 VV (3 × 106 pfu) and IL-2 (2 × 105 IU BID for 3 d) or no cell transfer (NT). Values are shown as means ± SEM *, P < 0.05 by two-way ANOVA. n = 5; two independent experiments. (C–E) IFN-γ evaluation by intracellular staining of patient PBL transduced with various tumor-specific TCRs, knocked down for CISH, and co-cultured with tumor for 6 h and assessed by flow cytometry. Retroviral transduced PBL with CISH shmiR or control shmiR and cotransduced with previously described TCR-encoding retroviruses directed against cancer-testis antigen, NY-ESO-1 (C), or MART-1 specific highly avid DMF5 (D) or poorly avid DMF4 TCR (E). 5 d after transduction, PBL were co-cultured with antigen-positive or -negative tumors and assessed for intracellular IFN-γ by flow cytometry. *, P < 0.05 by paired Student’s t test. n = 3–5; three independent experiments. (F) Transduced PBL from D were co-stained for IFN-γ, TNF, and IL-2 and evaluated for polycytokine functionality by flow cytometry and using a Boolean gating strategy. *, P < 0.05 by paired Student’s t test. n = 3; three independent experiments. (G) IFN-γ evaluation by ELISA of patient PBL knocked down for CISH and co-cultured overnight with relevant and irrelevant tumors. Retroviral transduced PBL with CISH shmiR or control shmiR and cotransduced with TCR-encoding retroviruses directed against cancer-testis antigens, MAGE-A1. 5 d after transduction, PBL were co-cultured with antigen-positive or -negative tumors. *, P < 0.05 by paired Student’s t test. n = 3; two independent experiments.
Figure 6.
Figure 6.
No overt changes in STAT5 activation in the absence of Cish at acute time points. (A) Western blot of Cish, phosphorylated STAT5, and β-actin in naive Cish−/− or WT pmel-1 T cells after αCD3 stimulation at times indicated; two independent experiments. (B) Schematic of STAT5 reporter self-inactivating (sin) retrovirus coexpressing the congenic marker Thy1.1 under the phospholgycerate kinase 1 promoter. (C) Relative luciferase activity in STAT5 reporter-transduced Cish−/− or WT-primed pmel-1 T cells after αCD3 stimulation at times indicated. Values are shown as means ± SEM. P > 0.05 by two-way ANOVA. n = 3; two independent experiments. (D) Flow cytometric evaluation of intracellular phosphorylated STAT5 in primed Cish−/− or WT pmel-1 T cells after the addition of IL-2 at indicated concentrations and times. P > 0.05 by two-way ANOVA. n = 3; three independent experiments.
Figure 7.
Figure 7.
TCR-dependent hyperactivation program in the absence of Cish. (A) Enhanced up-regulation of relative Tbx21, Cmyc, and Bcl2l1 gene expression by real-time PCR with or without αCD3 stimulation for 4 h. Values are shown as means ± SEM; n = 3. *, P < 0.05; **, P < 0.01 by two-way ANOVA. n = 3; two independent experiments. (B) Microarray volcano plot comparing in vitro–primed Cish−/− or WT CD8+ T cells 2 h after αCD3-stimulation. n = 3 independent mice per genotype; two independent experiments. (C and D) Gene-set enrichment analysis of genes in Cish-deficient T cells relative to WT littermates reveals a strong ranking with the Goldrath antigen response (C) and Kaech naive versus day 8 effector T cell down-regulated (D) profile.
Figure 8.
Figure 8.
Enhanced PLC-γ1 activation and downstream signaling in the absence of Cish. (A) Cish protein evaluation WT or Cish−/− CD8+ T cells with or without TCR stimulation by immunoblotting; three independent experiments. (B) Total phospho-Tyrosine (pTyr) blotting of cell lysates after αCD3 stimulation of Cish−/− or WT CD8+ T cells at times indicated by immunoblotting; three independent experiments. (C) Western blot analysis of phospho-PLC-γ1, total PLC-γ1, phosphor-LAT, whole LAT, phospho-Zap-70, whole Zap-70, and β-actin blot after αCD3 stimulation of Cish−/− or WT CD8+ T cells at times indicated; three independent experiments. (D) Representative Ca2+ flux as assessed by fluorometric evaluation after αCD3 stimulation of Cish−/− or WT CD8+ T cells. Kinetic of the ratio of Fluo3-AM by Fura Red over time shown and assessed by flow cytometry; five independent experiments. (E and F) Relative luciferase activity of NFAT and NF-κB reporter transduced Cish−/− or WT CD8+ T cells after αCD3 stimulation. ***, P < 0.001 by two-way ANOVA. n = 3; two independent experiments.
Figure 9.
Figure 9.
Cish specifically inhibits Ca2+ flux, T cell polyfunctionality, and PLC-γ1 accumulation in TCR microclusters. (A) Schematic of retroviral Cish expression vector. N-terminal Flag-tagged (3×) Cish, self-cleaving furin-2A (f2A) peptide, and congenic marker Thy1.1 driven by the intrinsic LTR promoter. (B) Representative Ca2+ flux as assessed by flow cytometry after αCD3 stimulation of Cish, empty control transduced Cish−/− CD8+ T cells, or empty control transduced WT CD8+ T cells. Kinetic of the ratio of Fluo3-AM by FuraRed over time shown and assessed by flow cytometry; four independent experiments. (C) Assessment of functional avidity by intracellular IFN-γ using flow cytometry. Cish or empty control transduced Cish−/− CD8+ T cells or empty control transduced WT CD8+ T cells from B were stimulated with indicated αCD3 concentrations for 6 h and evaluated by flow cytometry. Values represent mean fluorescence intensity. **, P < 0.01 by paired Student’s t test; three independent experiments. (D) Assessment of cytokine polyfunctionality in T cells from B using Boolean gating strategy. Data are shown as total percentage of two or three concomitant cytokines present after antigenic stimulation. n = 3; three independent experiments. (E and F) Evaluation of PLC-γ1 and phosphotyrosine in TCR microclusters after αCD3 stimulation. (E) Representative confocal images from transduced CD8+ T cells from B were dropped on to stimulatory coverslips, fixed after three minutes, immunostained for PLC-γ1 and phosphotyrosine (pTYR), and enumerated for intensity and area of each (F). Bars, 2 µm. ns, P > 0.05; **, P < 0.01 by unpaired Student’s t test. n = 18–24; three independent experiments.
Figure 10.
Figure 10.
TCR stimulation–dependent polyubiquitination of PLC-γ1 by Cish. (A) Immunoprecipitation of YFP-tagged PLC-γ1 and immunoblotting of Cish in transfected 293T cells in the absence of TCR signaling complex; two independent experiments. (B) Immunoprecipitation of FLAG-tagged Cish and immunoblotting of PLC-γ1 in indicated transduced CD8+ T cells with and without CD3 stimulation (5 min). Whole lysates were blotted for PLC-γ1 and Cish; two independent experiments. (C) Immunoprecipitation of endogenous Cish and immunoblotting of PLC-γ1 in 3 d Cish−/− or WT CD8+ T cell blasts; two independent experiments. (D) 293T cells were transfected with Tagged plasmids expressing PLC-γ1-YFP, Ubiquitin-HA, and Cish-FLAG where indicated in the presence of the proteasome inhibitor, MG-132. After transfection, PLC-γ1 was immunoprecipitated and blotted for HA and YFP. Whole-cell lysates were blotted for Cish and PLC-γ1-YFP; two independent experiments. (E) Immunoprecipitation of endogenous PLC-γ1 and immunoblotting of ubiquitin in indicated reconstituted CD8+ T cells with or without TCR stimulation in the presence of the proteasome inhibitor, MG-132; two independent experiments. (F) Immunoprecipitation of endogenous PLC-γ1 and immunoblotting of ubiquitin in native CD8+ T cells blasts (3 d) with or without TCR stimulation in the presence of the proteasome inhibitor, MG-132; two independent experiments.

References

    1. Acquavella N., Clever D., Yu Z., Roelke-Parker M., Palmer D.C., Xi L., Pflicke H., Ji Y., Gros A., Hanada K., et al. . 2015. Type I cytokines synergize with oncogene inhibition to induce tumor growth arrest. Cancer Immunol. Res. 3:37–47. 10.1158/2326-6066.CIR-14-0122
    1. Acuto O., Di Bartolo V., and Michel F.. 2008. Tailoring T-cell receptor signals by proximal negative feedback mechanisms. Nat. Rev. Immunol. 8:699–712. 10.1038/nri2397
    1. Aman M.J., Migone T.-S., Sasaki A., Ascherman D.P., Zhu Mh., Soldaini E., Imada K., Miyajima A., Yoshimura A., and Leonard W.J.. 1999. CIS associates with the interleukin-2 receptor beta chain and inhibits interleukin-2-dependent signaling. J. Biol. Chem. 274:30266–30272. 10.1074/jbc.274.42.30266
    1. Antony P.A., Piccirillo C.A., Akpinarli A., Finkelstein S.E., Speiss P.J., Surman D.R., Palmer D.C., Chan C.-C., Klebanoff C.A., Overwijk W.W., et al. . 2005. CD8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells. J. Immunol. 174:2591–2601. 10.4049/jimmunol.174.5.2591
    1. Arens R., and Schoenberger S.P.. 2010. Plasticity in programming of effector and memory CD8 T-cell formation. Immunol. Rev. 235:190–205.
    1. Babon J.J., McManus E.J., Yao S., DeSouza D.P., Mielke L.A., Sprigg N.S., Willson T.A., Hilton D.J., Nicola N.A., Baca M., et al. . 2006. The structure of SOCS3 reveals the basis of the extended SH2 domain function and identifies an unstructured insertion that regulates stability. Mol. Cell. 22:205–216. 10.1016/j.molcel.2006.03.024
    1. Balagopalan L., Ashwell B.A., Bernot K.M., Akpan I.O., Quasba N., Barr V.A., and Samelson L.E.. 2011. Enhanced T-cell signaling in cells bearing linker for activation of T-cell (LAT) molecules resistant to ubiquitylation. Proc. Natl. Acad. Sci. USA. 108:2885–2890. 10.1073/pnas.1007098108
    1. Batista F.D., and Dustin M.L.. 2013. Cell:cell interactions in the immune system. Immunol. Rev. 251:7–12. 10.1111/imr.12025
    1. Berg L.J., Finkelstein L.D., Lucas J.A., and Schwartzberg P.L.. 2005. Tec family kinases in T lymphocyte development and function. Annu. Rev. Immunol. 23:549–600. 10.1146/annurev.immunol.22.012703.104743
    1. Best J.A., Blair D.A., Knell J., Yang E., Mayya V., Doedens A., Dustin M.L., and Goldrath A.W.. Immunological Genome Project Consortium . 2013. Transcriptional insights into the CD8+ T cell response to infection and memory T cell formation. Nat. Immunol. 14:404–412. 10.1038/ni.2536
    1. Braiman A., Barda-Saad M., Sommers C.L., and Samelson L.E.. 2006. Recruitment and activation of PLCgamma1 in T cells: a new insight into old domains. EMBO J. 25:774–784. 10.1038/sj.emboj.7600978
    1. Bunnell S.C., Hong D.I., Kardon J.R., Yamazaki T., McGlade C.J., Barr V.A., and Samelson L.E.. 2002. T cell receptor ligation induces the formation of dynamically regulated signaling assemblies. J. Cell Biol. 158:1263–1275. 10.1083/jcb.200203043
    1. Catlett I.M., and Hedrick S.M.. 2005. Suppressor of cytokine signaling 1 is required for the differentiation of CD4+ T cells. Nat. Immunol. 6:715–721. 10.1038/ni1211
    1. Chaigne-Delalande B., Li F.Y., O’Connor G.M., Lukacs M.J., Jiang P., Zheng L., Shatzer A., Biancalana M., Pittaluga S., Matthews H.F., et al. . 2013. Mg2+ regulates cytotoxic functions of NK and CD8 T cells in chronic EBV infection through NKG2D. Science. 341:186–191. 10.1126/science.1240094
    1. Chervin A.S., Stone J.D., Soto C.M., Engels B., Schreiber H., Roy E.J., and Kranz D.M.. 2013. Design of T-cell receptor libraries with diverse binding properties to examine adoptive T-cell responses. Gene Ther. 20:634–644. 10.1038/gt.2012.80
    1. Cohney S.J., Sanden D., Cacalano N.A., Yoshimura A., Mui A., Migone T.S., and Johnston J.A.. 1999. SOCS-3 is tyrosine phosphorylated in response to interleukin-2 and suppresses STAT5 phosphorylation and lymphocyte proliferation. Mol. Cell. Biol. 19:4980–4988.
    1. Davey G.M., Starr R., Cornish A.L., Burghardt J.T., Alexander W.S., Carbone F.R., Surh C.D., and Heath W.R.. 2005. SOCS-1 regulates IL-15-driven homeostatic proliferation of antigen-naive CD8 T cells, limiting their autoimmune potential. J. Exp. Med. 202:1099–1108. 10.1084/jem.20050003
    1. Davis J.L., Theoret M.R., Zheng Z., Lamers C.H.J., Rosenberg S.A., and Morgan R.A.. 2010. Development of human anti-murine T-cell receptor antibodies in both responding and nonresponding patients enrolled in TCR gene therapy trials. Clin. Cancer Res. 16:5852–5861. 10.1158/1078-0432.CCR-10-1280
    1. Dif F., Saunier E., Demeneix B., Kelly P.A., and Edery M.. 2001. Cytokine-inducible SH2-containing protein suppresses PRL signaling by binding the PRL receptor. Endocrinology. 142:5286–5293. 10.1210/endo.142.12.8549
    1. Dissanayake D., Gronski M.A., Lin A., Elford A.R., and Ohashi P.S.. 2011. Immunological perspective of self versus tumor antigens: insights from the RIP-gp model. Immunol. Rev. 241:164–179. 10.1111/j.1600-065X.2011.01014.x
    1. Dranoff G. 2013. Immunotherapy at large: Balancing tumor immunity and inflammatory pathology. Nat. Med. 19:1100–1101. 10.1038/nm.3335
    1. Dranoff G., and Fearon D.. 2013. Tumour immunology. Curr. Opin. Immunol. 25:189–191. 10.1016/j.coi.2013.02.012
    1. Dudda J.C., Salaun B., Ji Y., Palmer D.C., Monnot G.C., Merck E., Boudousquie C., Utzschneider D.T., Escobar T.M., Perret R., et al. . 2013. MicroRNA-155 is required for effector CD8+ T cell responses to virus infection and cancer. Immunity. 38:742–753. 10.1016/j.immuni.2012.12.006
    1. Endo T., Sasaki A., Minoguchi M., Joo A., and Yoshimura A.. 2003. CIS1 interacts with the Y532 of the prolactin receptor and suppresses prolactin-dependent STAT5 activation. J. Biochem. 133:109–113. 10.1093/jb/mvg004
    1. Endo T.A., Masuhara M., Yokouchi M., Suzuki R., Sakamoto H., Mitsui K., Matsumoto A., Tanimura S., Ohtsubo M., Misawa H., et al. . 1997. A new protein containing an SH2 domain that inhibits JAK kinases. Nature. 387:921–924. 10.1038/43213
    1. Gajewski T.F., Schreiber H., and Fu Y.-X.. 2013. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14:1014–1022. 10.1038/ni.2703
    1. Goldrath A.W., Luckey C.J., Park R., Benoist C., and Mathis D.. 2004. The molecular program induced in T cells undergoing homeostatic proliferation. Proc. Natl. Acad. Sci. USA. 101:16885–16890. 10.1073/pnas.0407417101
    1. Gronski M.A., Boulter J.M., Moskophidis D., Nguyen L.T., Holmberg K., Elford A.R., Deenick E.K., Kim H.O., Penninger J.M., Odermatt B., et al. . 2004. TCR affinity and negative regulation limit autoimmunity. Nat. Med. 10:1234–1239. 10.1038/nm1114
    1. Guittard G., Kortum R.L., Balagopalan L., Çuburu N., Nguyen P., Sommers C.L., and Samelson L.E.. 2015. Absence of both Sos-1 and Sos-2 in peripheral CD4+ T cells leads to PI3K pathway activation and defects in migration. Eur. J. Immunol. 45:2389–2395. 10.1002/eji.201445226
    1. Harlin H., Kuna T.V., Peterson A.C., Meng Y., and Gajewski T.F.. 2006. Tumor progression despite massive influx of activated CD8(+) T cells in a patient with malignant melanoma ascites. Cancer Immunol. Immunother. 55:1185–1197. 10.1007/s00262-005-0118-2
    1. Hilton D.J., Richardson R.T., Alexander W.S., Viney E.M., Willson T.A., Sprigg N.S., Starr R., Nicholson S.E., Metcalf D., and Nicola N.A.. 1998. Twenty proteins containing a C-terminal SOCS box form five structural classes. Proc. Natl. Acad. Sci. USA. 95:114–119. 10.1073/pnas.95.1.114
    1. Hodi F.S., and Dranoff G.. 2010. The biologic importance of tumor-infiltrating lymphocytes. J. Cutan. Pathol. 37:48–53. 10.1111/j.1600-0560.2010.01506.x
    1. Janicki C.N., Jenkinson S.R., Williams N.A., and Morgan D.J.. 2008. Loss of CTL function among high-avidity tumor-specific CD8+ T cells following tumor infiltration. Cancer Res. 68:2993–3000. 10.1158/0008-5472.CAN-07-5008
    1. Ji Y., Pos Z., Rao M., Klebanoff C.A., Yu Z., Sukumar M., Reger R.N., Palmer D.C., Borman Z.A., Muranski P., et al. . 2011. Repression of the DNA-binding inhibitor Id3 by Blimp-1 limits the formation of memory CD8+ T cells. Nat. Immunol. 12:1230–1237. 10.1038/ni.2153
    1. Jin P., Wang E., Provenzano M., Deola S., Selleri S., Ren J., Voiculescu S., Stroncek D., Panelli M.C., and Marincola F.M.. 2006. Molecular signatures induced by interleukin-2 on peripheral blood mononuclear cells and T cell subsets. J. Transl. Med. 4:26 10.1186/1479-5876-4-26
    1. Johnson L.A., Heemskerk B., Powell D.J. Jr., Cohen C.J., Morgan R.A., Dudley M.E., Robbins P.F., and Rosenberg S.A.. 2006. Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes. J. Immunol. 177:6548–6559. 10.4049/jimmunol.177.9.6548
    1. June C.H., Fletcher M.C., Ledbetter J.A., Schieven G.L., Siegel J.N., Phillips A.F., and Samelson L.E.. 1990. Inhibition of tyrosine phosphorylation prevents T-cell receptor-mediated signal transduction. Proc. Natl. Acad. Sci. USA. 87:7722–7726. 10.1073/pnas.87.19.7722
    1. Kaech S.M., Hemby S., Kersh E., and Ahmed R.. 2002a Molecular and functional profiling of memory CD8 T cell differentiation. Cell. 111:837–851. 10.1016/S0092-8674(02)01139-X
    1. Kaech S.M., Wherry E.J., and Ahmed R.. 2002b Effector and memory T-cell differentiation: implications for vaccine development. Nat. Rev. Immunol. 2:251–262. 10.1038/nri778
    1. Kalos M., and June C.H.. 2013. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity. 39:49–60. 10.1016/j.immuni.2013.07.002
    1. Kamizono S., Hanada T., Yasukawa H., Minoguchi S., Kato R., Minoguchi M., Hattori K., Hatakeyama S., Yada M., Morita S., et al. . 2001. The SOCS box of SOCS-1 accelerates ubiquitin-dependent proteolysis of TEL-JAK2. J. Biol. Chem. 276:12530–12538. 10.1074/jbc.M010074200
    1. Kerkar S.P., Muranski P., Kaiser A., Boni A., Sanchez-Perez L., Yu Z., Palmer D.C., Reger R.N., Borman Z.A., Zhang L., et al. . 2010. Tumor-specific CD8+ T cells expressing interleukin-12 eradicate established cancers in lymphodepleted hosts. Cancer Res. 70:6725–6734. 10.1158/0008-5472.CAN-10-0735
    1. Khor C.C., Vannberg F.O., Chapman S.J., Guo H., Wong S.H., Walley A.J., Vukcevic D., Rautanen A., Mills T.C., Chang K.C., et al. . 2010. CISH and susceptibility to infectious diseases. N. Engl. J. Med. 362:2092–2101. 10.1056/NEJMoa0905606
    1. King C.G., Koehli S., Hausmann B., Schmaler M., Zehn D., and Palmer E.. 2012. T cell affinity regulates asymmetric division, effector cell differentiation, and tissue pathology. Immunity. 37:709–720. 10.1016/j.immuni.2012.06.021
    1. Landsman T., and Waxman D.J.. 2005. Role of the cytokine-induced SH2 domain-containing protein CIS in growth hormone receptor internalization. J. Biol. Chem. 280:37471–37480. 10.1074/jbc.M504125200
    1. Li S., Chen S., Xu X., Sundstedt A., Paulsson K.M., Anderson P., Karlsson S., Sjögren H.O., and Wang P.. 2000. Cytokine-induced Src homology 2 protein (CIS) promotes T cell receptor-mediated proliferation and prolongs survival of activated T cells. J. Exp. Med. 191:985–994. 10.1084/jem.191.6.985
    1. Linette G.P., Stadtmauer E.A., Maus M.V., Rapoport A.P., Levine B.L., Emery L., Litzky L., Bagg A., Carreno B.M., Cimino P.J., et al. . 2013. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 122:863–871. 10.1182/blood-2013-03-490565
    1. Malecek K., Zhong S., McGary K., Yu C., Huang K., Johnson L.A., Rosenberg S.A., and Krogsgaard M.. 2013. Engineering improved T cell receptors using an alanine-scan guided T cell display selection system. J. Immunol. Methods. 392:1–11. 10.1016/j.jim.2013.02.018
    1. Manicassamy S., Gupta S., Huang Z., and Sun Z.. 2006. Protein kinase C-theta-mediated signals enhance CD4+ T cell survival by up-regulating Bcl-xL. J. Immunol. 176:6709–6716. 10.4049/jimmunol.176.11.6709
    1. Matsumoto A., Masuhara M., Mitsui K., Yokouchi M., Ohtsubo M., Misawa H., Miyajima A., and Yoshimura A.. 1997. CIS, a cytokine inducible SH2 protein, is a target of the JAK-STAT5 pathway and modulates STAT5 activation. Blood. 89:3148–3154.
    1. Maus M.V., Fraietta J.A., Levine B.L., Kalos M., Zhao Y., and June C.H.. 2014. Adoptive immunotherapy for cancer or viruses. Annu. Rev. Immunol. 32:189–225. 10.1146/annurev-immunol-032713-120136
    1. Morgan R.A., Chinnasamy N., Abate-Daga D., Gros A., Robbins P.F., Zheng Z., Dudley M.E., Feldman S.A., Yang J.C., Sherry R.M., et al. . 2013. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J. Immunother. 36:133–151. 10.1097/CJI.0b013e3182829903
    1. Mortarini R., Piris A., Maurichi A., Molla A., Bersani I., Bono A., Bartoli C., Santinami M., Lombardo C., Ravagnani F., et al. . 2003. Lack of terminally differentiated tumor-specific CD8+ T cells at tumor site in spite of antitumor immunity to self-antigens in human metastatic melanoma. Cancer Res. 63:2535–2545.
    1. Naka T., Narazaki M., Hirata M., Matsumoto T., Minamoto S., Aono A., Nishimoto N., Kajita T., Taga T., Yoshizaki K., et al. . 1997. Structure and function of a new STAT-induced STAT inhibitor. Nature. 387:924–929. 10.1038/43219
    1. Nauerth M., Weißbrich B., Knall R., Franz T., Dössinger G., Bet J., Paszkiewicz P.J., Pfeifer L., Bunse M., Uckert W., et al. . 2013. TCR-ligand koff rate correlates with the protective capacity of antigen-specific CD8+ T cells for adoptive transfer. Sci. Transl. Med. 5:192ra87.
    1. Ohashi P.S., Oehen S., Buerki K., Pircher H., Ohashi C.T., Odermatt B., Malissen B., Zinkernagel R.M., and Hengartner H.. 1991. Ablation of “tolerance” and induction of diabetes by virus infection in viral antigen transgenic mice. Cell. 65:305–317.
    1. Overwijk W.W., Theoret M.R., Finkelstein S.E., Surman D.R., de Jong L.A., Vyth-Dreese F.A., Dellemijn T.A., Antony P.A., Spiess P.J., Palmer D.C., et al. . 2003. Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells. J. Exp. Med. 198:569–580. 10.1084/jem.20030590
    1. Palmer D.C., and Restifo N.P.. 2009. Suppressors of cytokine signaling (SOCS) in T cell differentiation, maturation, and function. Trends Immunol. 30:592–602. 10.1016/j.it.2009.09.009
    1. Palmer D.C., Chan C.C., Gattinoni L., Wrzesinski C., Paulos C.M., Hinrichs C.S., Powell D.J. Jr., Klebanoff C.A., Finkelstein S.E., Fariss R.N., et al. . 2008. Effective tumor treatment targeting a melanoma/melanocyte-associated antigen triggers severe ocular autoimmunity. Proc. Natl. Acad. Sci. USA. 105:8061–8066. 10.1073/pnas.0710929105
    1. Rabinovich G.A., Gabrilovich D., and Sotomayor E.M.. 2007. Immunosuppressive strategies that are mediated by tumor cells. Annu. Rev. Immunol. 25:267–296. 10.1146/annurev.immunol.25.022106.141609
    1. Rao M., Chinnasamy N., Hong J.A., Zhang Y., Zhang M., Xi S., Liu F., Marquez V.E., Morgan R.A., and Schrump D.S.. 2011. Inhibition of histone lysine methylation enhances cancer-testis antigen expression in lung cancer cells: implications for adoptive immunotherapy of cancer. Cancer Res. 71:4192–4204. 10.1158/0008-5472.CAN-10-2442
    1. Restifo N.P., Dudley M.E., and Rosenberg S.A.. 2012. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat. Rev. Immunol. 12:269–281. 10.1038/nri3191
    1. Rosati S.F., Parkhurst M.R., Hong Y., Zheng Z., Feldman S.A., Rao M., Abate-Daga D., Beard R.E., Xu H., Black M.A., et al. . 2014. A novel murine T-cell receptor targeting NY-ESO-1. J. Immunother. 37:135–146. 10.1097/CJI.0000000000000019
    1. Rosenberg S.A., Sherry R.M., Morton K.E., Scharfman W.J., Yang J.C., Topalian S.L., Royal R.E., Kammula U., Restifo N.P., Hughes M.S., et al. . 2005. Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma. J. Immunol. 175:6169–6176. 10.4049/jimmunol.175.9.6169
    1. Scholer A., Hugues S., Boissonnas A., Fetler L., and Amigorena S.. 2008. Intercellular adhesion molecule-1-dependent stable interactions between T cells and dendritic cells determine CD8+ T cell memory. Immunity. 28:258–270. 10.1016/j.immuni.2007.12.016
    1. Seki Y., Inoue H., Nagata N., Hayashi K., Fukuyama S., Matsumoto K., Komine O., Hamano S., Himeno K., Inagaki-Ohara K., et al. . 2003. SOCS-3 regulates onset and maintenance of T(H)2-mediated allergic responses. Nat. Med. 9:1047–1054. 10.1038/nm896
    1. Smith-Garvin J.E., Koretzky G.A., and Jordan M.S.. 2009. T cell activation. Annu. Rev. Immunol. 27:591–619. 10.1146/annurev.immunol.021908.132706
    1. Sommers C.L., Lee J., Steiner K.L., Gurson J.M., Depersis C.L., El-Khoury D., Fuller C.L., Shores E.W., Love P.E., and Samelson L.E.. 2005. Mutation of the phospholipase C-gamma1-binding site of LAT affects both positive and negative thymocyte selection. J. Exp. Med. 201:1125–1134. 10.1084/jem.20041869
    1. Starr R., Willson T.A., Viney E.M., Murray L.J., Rayner J.R., Jenkins B.J., Gonda T.J., Alexander W.S., Metcalf D., Nicola N.A., and Hilton D.J.. 1997. A family of cytokine-inducible inhibitors of signalling. Nature. 387:917–921. 10.1038/43206
    1. Taleb S., Romain M., Ramkhelawon B., Uyttenhove C., Pasterkamp G., Herbin O., Esposito B., Perez N., Yasukawa H., Van Snick J., et al. . 2009. Loss of SOCS3 expression in T cells reveals a regulatory role for interleukin-17 in atherosclerosis. J. Exp. Med. 206:2067–2077. 10.1084/jem.20090545
    1. Tanaka K., Ichiyama K., Hashimoto M., Yoshida H., Takimoto T., Takaesu G., Torisu T., Hanada T., Yasukawa H., Fukuyama S., et al. . 2008. Loss of suppressor of cytokine signaling 1 in helper T cells leads to defective Th17 differentiation by enhancing antagonistic effects of IFN-gamma on STAT3 and Smads. J. Immunol. 180:3746–3756. 10.4049/jimmunol.180.6.3746
    1. Thaventhiran J.E., Fearon D.T., and Gattinoni L.. 2013. Transcriptional regulation of effector and memory CD8+ T cell fates. Curr. Opin. Immunol. 25:321–328. 10.1016/j.coi.2013.05.010
    1. Tong H.V., Toan N.L., Song H., Kremsner P.G., Kun J.F., and Tp V.. 2012. Association of CISH-292A/T genetic variant with hepatitis B virus infection. Immunogenetics. 64:261–265. 10.1007/s00251-011-0584-y
    1. Uyttendaele I., Lemmens I., Verhee A., De Smet A.S., Vandekerckhove J., Lavens D., Peelman F., and Tavernier J.. 2007. Mammalian protein-protein interaction trap (MAPPIT) analysis of STAT5, CIS, and SOCS2 interactions with the growth hormone receptor. Mol. Endocrinol. 21:2821–2831. 10.1210/me.2006-0541
    1. Vazquez-Cintron E.J., Monu N.R., and Frey A.B.. 2010. Tumor-induced disruption of proximal TCR-mediated signal transduction in tumor-infiltrating CD8+ lymphocytes inactivates antitumor effector phase. J. Immunol. 185:7133–7140. 10.4049/jimmunol.1001157
    1. Whiteside T.L. 2006. Immune suppression in cancer: effects on immune cells, mechanisms and future therapeutic intervention. Semin. Cancer Biol. 16:3–15. 10.1016/j.semcancer.2005.07.008
    1. Yang X.O., Zhang H., Kim B.S., Niu X., Peng J., Chen Y., Kerketta R., Lee Y.H., Chang S.H., Corry D.B., et al. . 2013. The signaling suppressor CIS controls proallergic T cell development and allergic airway inflammation. Nat. Immunol. 14:732–740. 10.1038/ni.2633
    1. Yasukawa H., Sasaki A., and Yoshimura A.. 2000. Negative regulation of cytokine signaling pathways. Annu. Rev. Immunol. 18:143–164. 10.1146/annurev.immunol.18.1.143
    1. Yokosuka T., Sakata-Sogawa K., Kobayashi W., Hiroshima M., Hashimoto-Tane A., Tokunaga M., Dustin M.L., and Saito T.. 2005. Newly generated T cell receptor microclusters initiate and sustain T cell activation by recruitment of Zap70 and SLP-76. Nat. Immunol. 6:1253–1262. 10.1038/ni1272
    1. Yoshimura A. 2013. CIS: the late-blooming eldest son. Nat. Immunol. 14:692–694. 10.1038/ni.2645
    1. Yoshimura A., Ohkubo T., Kiguchi T., Jenkins N.A., Gilbert D.J., Copeland N.G., Hara T., and Miyajima A.. 1995. A novel cytokine-inducible gene CIS encodes an SH2-containing protein that binds to tyrosine-phosphorylated interleukin 3 and erythropoietin receptors. EMBO J. 14:2816–2826.
    1. Yoshimura A., Naka T., and Kubo M.. 2007. SOCS proteins, cytokine signalling and immune regulation. Nat. Rev. Immunol. 7:454–465. 10.1038/nri2093
    1. Zhang N., and Bevan M.J.. 2011. CD8+ T cells: foot soldiers of the immune system. Immunity. 35:161–168. 10.1016/j.immuni.2011.07.010
    1. Zhang J.G., Farley A., Nicholson S.E., Willson T.A., Zugaro L.M., Simpson R.J., Moritz R.L., Cary D., Richardson R., Hausmann G., et al. . 1999. The conserved SOCS box motif in suppressors of cytokine signaling binds to elongins B and C and may couple bound proteins to proteasomal degradation. Proc. Natl. Acad. Sci. USA. 96:2071–2076. 10.1073/pnas.96.5.2071
    1. Zhang L., Feldman S.A., Zheng Z., Chinnasamy N., Xu H., Nahvi A.V., Dudley M.E., Rosenberg S.A., and Morgan R.A.. 2012. Evaluation of γ-retroviral vectors that mediate the inducible expression of IL-12 for clinical application. J. Immunother. 35:430–439. 10.1097/CJI.0b013e31825898e8
    1. Zhao Y., Bennett A.D., Zheng Z., Wang Q.J., Robbins P.F., Yu L.Y.L., Li Y., Molloy P.E., Dunn S.M., Jakobsen B.K., et al. . 2007. High-affinity TCRs generated by phage display provide CD4+ T cells with the ability to recognize and kill tumor cell lines. J. Immunol. 179:5845–5854. 10.4049/jimmunol.179.9.5845
    1. Zippelius A., Batard P., Rubio-Godoy V., Bioley G., Liénard D., Lejeune F., Rimoldi D., Guillaume P., Meidenbauer N., Mackensen A., et al. . 2004. Effector function of human tumor-specific CD8 T cells in melanoma lesions: a state of local functional tolerance. Cancer Res. 64:2865–2873. 10.1158/0008-5472.CAN-03-3066

Source: PubMed

3
Prenumerera