J. Koch, A. Steinle, C. Watzl, and O. Mandelboim, Activating natural cytotoxicity receptors of natural killer cells in cancer and infection, Trends Immunol, vol.34, pp.182-191, 2013.

T. Pazina, A. Shemesh, M. Brusilovsky, A. Porgador, and K. S. Campbell, Regulation of the functions of natural cytotoxicity receptors by interactions with diverse ligands and alterations in splice variant expression, Front Immunol, vol.8, p.369, 2017.

C. S. Brandt, M. Baratin, E. C. Yi, J. Kennedy, Z. Gao et al., The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans, J Exp Med, vol.206, pp.1495-1503, 2009.

G. Cao, J. Wang, X. Zheng, H. Wei, Z. Tian et al., Tumor therapeutics work as stress inducers to enhance tumor sensitivity to Natural Killer (NK) cell cytolysis by up-regulating NKp30 ligand B7-H6, J Biol Chem, vol.290, pp.29964-29973, 2015.

M. Salimi, L. Xue, H. Jolin, C. Hardman, D. J. Cousins et al., Group 2 innate lymphoid cells express functional NKp30 receptor inducing type 2 cytokine production, J Immunol, vol.196, pp.45-54, 2016.

J. Matta, M. Baratin, L. Chiche, J. M. Forel, C. Cognet et al., Induction of B7-H6, a ligand for the natural killer cell-activating receptor NKp30, in inflammatory conditions, Blood, vol.122, pp.394-404, 2013.

C. A. Fielding, M. P. Weekes, L. V. Nobre, E. Ruckova, G. S. Wilkie et al., Control of immune ligands by members of a cytomegalovirus gene expansion suppresses natural killer cell activation, 2017.

Y. Charpak-amikam, T. Kubsch, E. Seidel, E. Oiknine-djian, N. Cavaletto et al., Human cytomegalovirus escapes immune recognition by NK cells through the downregulation of B7-H6 by the viral genes US18 and US20, Sci Rep, vol.7, p.8661, 2017.

D. Schmiedel, J. Tai, F. Levi-schaffer, S. Dovrat, and O. Mandelboim, Human Herpesvirus 6B Downregulates expression of activating ligands during lytic infection to escape elimination by natural killer cells, J Virol, vol.90, pp.9608-9617, 2016.

O. Lucar, S. Diallo, M. Bayard, C. Samri, A. Tarantino et al., B7-H6-mediated downregulation of NKp30 in natural killer cells contributes to HIV-2 immune escape, AIDS, vol.33, pp.23-32, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02307134

M. R. Wu, T. Zhang, L. R. Demars, and C. L. Sentman, B7H6-specific chimeric antigen receptors lead to tumor elimination and host antitumor immunity, Gene Ther, vol.22, pp.675-684, 2015.

M. R. Wu, T. Zhang, A. T. Gacerez, T. A. Coupet, L. R. Demars et al., B7H6-specific bispecific T cell engagers Lead to tumor elimination and host antitumor immunity, J Immunol, vol.194, pp.5305-5311, 2015.

S. Textor, F. Bossler, K. O. Henrich, M. Gartlgruber, J. Pollmann et al., The proto-oncogene Myc drives expression of the NK cell-activating NKp30 ligand B7-H6 in tumor cells, Oncoimmunology, vol.5, p.1116674, 2016.

E. Xia, Y. Shen, A. Bhandari, X. Zhou, Y. Wang et al., Long non-coding RNA LINC00673 promotes breast cancer proliferation and metastasis through regulating B7-H6 and epithelial-mesenchymal transition, Am J Cancer Res, vol.8, pp.1273-1287, 2018.

C. Hetz, E. Chevet, and S. A. Oakes, Proteostasis control by the unfolded protein response, Nat Cell Biol, vol.17, pp.829-838, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01175531

D. Ron and P. Walter, Signal integration in the endoplasmic reticulum unfolded protein response, Nat Rev Mol Cell Biol, vol.8, pp.519-529, 2007.

K. Pakos-zebrucka, I. Koryga, K. Mnich, M. Ljujic, A. Samali et al., The integrated stress response, EMBO Rep, vol.17, pp.1374-1395, 2016.

H. P. Harding, I. Novoa, Y. Zhang, H. Zeng, R. Wek et al., Regulated translation initiation controls stress-induced gene expression in mammalian cells, Mol Cell, vol.6, pp.1099-1108, 2000.

N. Gonen, N. Sabath, C. B. Burge, and R. Shalgi, Widespread PERK-dependent repression of ER targets in response to ER stress, Sci Rep, vol.9, p.4330, 2019.

N. N. Iwakoshi, A. H. Lee, P. Vallabhajosyula, K. L. Otipoby, K. Rajewsky et al., Plasma cell differentiation and the unfolded protein response intersect at the transcription factor XBP-1, Nat Immunol, vol.4, pp.321-329, 2003.

J. Pramanik, X. Chen, G. Kar, J. Henriksson, T. Gomes et al., , 2018.

, Genome-wide analyses reveal the IRE1a-XBP1 pathway promotes T helper cell differentiation by resolving secretory stress and accelerating proliferation, Genome Med, vol.10, p.76

M. Wang and R. J. Kaufman, The impact of the endoplasmic reticulum protein-folding environment on cancer development, Nat Rev Cancer, vol.14, pp.581-597, 2014.

S. Hosomi, J. Grootjans, M. Tschurtschenthaler, N. Krupka, J. D. Matute et al., Intestinal epithelial cell endoplasmic reticulum stress promotes MULT1 up-regulation and NKG2D-mediated inflammation, J Exp Med, vol.214, pp.2985-2997, 2017.

A. Obiedat, E. Seidel, M. Mahameed, O. Berhani, P. Tsukerman et al., Transcription of the NKG2D ligand MICA is suppressed by the IRE1/XBP1 pathway of the unfolded protein response through the regulation of E2F1, FASEB J, vol.33, pp.3481-3495, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01940305

A. Bercovich-kinori, J. Tai, I. A. Gelbart, A. Shitrit, S. Ben-moshe et al., A systematic view on influenza induced host shutoff, 2016.

J. M. Axten, J. R. Medina, Y. Feng, A. Shu, S. P. Romeril et al., Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-p yrrolo[2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), J Med Chem, vol.55, pp.7193-7207, 2012.

C. Sidrauski, D. Acosta-alvear, A. Khoutorsky, P. Vedantham, B. R. Hearn et al., Pharmacological brake-release of mRNA translation enhances cognitive memory, vol.2, p.498, 2013.

B. J. Guan, V. Van-hoef, R. Jobava, O. Elroy-stein, L. S. Valasek et al., A unique ISR program determines cellular responses to chronic stress, Mol Cell, vol.68, p.886, 2017.

M. Siddiquey, H. Zhang, C. C. Nguyen, A. J. Domma, and J. P. Kamil, The human cytomegalovirus endoplasmic reticulumresident glycoprotein UL148 activates the unfolded protein response, J Virol, vol.92, 2018.

P. Tsaytler, H. P. Harding, R. D. Bertolotti, and A. , Selective inhibition of a regulatory subunit of protein phosphatase 1 restores proteostasis, Science, vol.332, pp.91-94, 2011.

W. B'chir, A. C. Maurin, V. Carraro, J. Averous, C. Jousse et al., The eIF2alpha/ATF4 pathway is essential for stress-induced autophagy gene expression, Nucleic Acids Res, vol.41, pp.7683-7699, 2013.

D. Gassart, A. Bujisic, B. Zaffalon, L. Decosterd, L. A. et al., An inhibitor of HIV-1 protease modulates constitutive eIF2alpha dephosphorylation to trigger a specific integrated stress response, Proc Natl Acad Sci U S A, vol.113, pp.117-126, 2016.

R. Liu, L. Zhang, J. Yang, X. Zhang, R. Mikkelsen et al., HIV protease inhibitors sensitize human head and neck squamous carcinoma cells to radiation by activating endoplasmic reticulum stress, PLoS One, vol.10, p.125928, 2015.

Y. J. Bryson, M. Mirochnick, A. Stek, L. M. Mofenson, J. Connor et al., Pharmacokinetics and safety of nelfinavir when used in combination with zidovudine and lamivudine in HIV-infected pregnant women: pediatric AIDS Clinical Trials Group (PACTG) protocol 353, HIV Clin Trials, vol.9, pp.115-125, 2008.

G. M. Blumenthal, J. J. Gills, M. S. Ballas, W. B. Bernstein, T. Komiya et al., A phase I trial of the HIV protease inhibitor nelfinavir in adults with solid tumors, Oncotarget, vol.5, pp.8161-8172, 2014.

C. Driessen, M. Kraus, M. Joerger, H. Rosing, J. Bader et al., Treatment with the HIV protease inhibitor nelfinavir triggers the unfolded protein response and may overcome proteasome inhibitor resistance of multiple myeloma in combination with bortezomib: a phase I trial (SAKK 65/08), Haematologica, vol.101, pp.346-355, 2016.

C. Franklin, E. Livingstone, A. Roesch, B. Schilling, and D. Schadendorf, Immunotherapy in melanoma: recent advances and future directions, Eur J Surg Oncol, vol.43, pp.604-611, 2017.

P. Sharma and J. P. Allison, The future of immune checkpoint therapy, Science, vol.348, pp.56-61, 2015.

L. S. Hart, J. T. Cunningham, T. Datta, S. Dey, F. Tameire et al., ER stress-mediated autophagy promotes Myc-dependent transformation and tumor growth, J Clin Invest, vol.122, pp.4621-4634, 2012.

Z. Shi, X. Yu, M. Yuan, W. Lv, T. Feng et al., Activation of the PERK-ATF4 pathway promotes chemoresistance in colon cancer cells, Sci Rep, vol.9, p.3210, 2019.

D. Pytel, Y. Gao, K. Mackiewicz, Y. V. Katlinskaya, K. A. Staschke et al., PERK is a Haploinsufficient tumor suppressor: gene dose determines tumor-suppressive versus tumor promoting properties of PERK in melanoma, PLoS Genet, vol.12, p.1006518, 2016.

L. R. Palam, T. D. Baird, and R. C. Wek, Phosphorylation of eIF2 facilitates ribosomal bypass of an inhibitory upstream ORF to enhance CHOP translation, J Biol Chem, vol.286, pp.10939-10949, 2011.

P. D. Lu, H. P. Harding, and R. D. , Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response, J Cell Biol, vol.167, pp.27-33, 2004.

S. R. Starck, J. C. Tsai, K. Chen, M. Shodiya, L. Wang et al., Translation from the 5' untranslated region shapes the integrated stress response, Science, vol.351, p.3867, 2016.

X. Wu, L. Sun, W. Zha, E. Studer, E. Gurley et al., HIV protease inhibitors induce endoplasmic reticulum stress and disrupt barrier integrity in intestinal epithelial cells, Gastroenterology, vol.138, pp.197-209, 2010.

S. F. Wang, M. S. Chen, Y. C. Chou, Y. F. Ueng, P. H. Yin et al., Mitochondrial dysfunction enhances cisplatin resistance in human gastric cancer cells via the ROS-activated GCN2-eIF2alpha-ATF4-xCT pathway, Oncotarget, vol.7, pp.74132-74151, 2016.

M. Abdel-nour, L. Carneiro, J. Downey, J. Tsalikis, A. Outlioua et al., The heme-regulated inhibitor is a cytosolic sensor of protein misfolding that controls innate immune signaling, Science, vol.365, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02372326

N. N. Shah, T. Maatman, P. Hari, and B. Johnson, Multi targeted CAR-T cell therapies for B-cell malignancies, Front Oncol, vol.9, p.146, 2019.

S. S. Neelapu, F. L. Locke, N. L. Bartlett, L. J. Lekakis, D. B. Miklos et al., Axicabtagene Ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma, N Engl J Med, vol.377, pp.2531-2544, 2017.

N. N. Shah and T. J. Fry, Mechanisms of resistance to CAR T cell therapy, Nat Rev Clin Oncol, vol.16, pp.372-385, 2019.

, Publisher's note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations