Skip to main content
Top
Published in: Cellular Oncology 4/2020

01-08-2020 | Solid Tumor | Review

Functional and metabolic targeting of natural killer cells to solid tumors

Authors: Jiao Wang, Sandro Matosevic

Published in: Cellular Oncology | Issue 4/2020

Login to get access

Abstract

Background

The unique ability of NK cells to target cancer cells without antigen specificity makes them an attractive prospect for immunotherapy of solid tumors. However, the complexity of the tumor microenvironment (TME), particularly its heterogeneity and associated immunosuppressive properties, enables solid tumor cells to escape NK cell immune-surveillance by impairing their infiltration and cytotoxic functions. As a result, NK cells that have been able to infiltrate solid tumors are dysfunctional, exhausted and metabolically and functionally impaired. Understanding the status of NK cells in solid tumors and the interplay between the tumor-promoting functions of the TME and the immunometabolic reprogramming events that NK cells endure as a result is essential to developing approaches to improve the clinical outcome of NK cell-based immunotherapies against solid tumors.

Conclusions

In this review, we address the current knowledge on the presence and immunometabolic roles of NK cells in solid tumors as well as the strategies developed to restore NK cell activities in these conditions, with the ultimate goal of enhancing persistence, trafficking, cytotoxicity and metabolic functions.
Literature
1.
go back to reference C. Guillerey, N.D. Huntington, M.J. Smyth, Targeting natural killer cells in cancer immunotherapy. Nat Immunol 17, 1025–1036 (2016)PubMed C. Guillerey, N.D. Huntington, M.J. Smyth, Targeting natural killer cells in cancer immunotherapy. Nat Immunol 17, 1025–1036 (2016)PubMed
2.
go back to reference A.M. Abel, C. Yang, M.S. Thakar, S. Malarkannan, Natural killer cells: Development, maturation, and clinical utilization. Front Immunol 9, 1869 (2018)PubMedPubMedCentral A.M. Abel, C. Yang, M.S. Thakar, S. Malarkannan, Natural killer cells: Development, maturation, and clinical utilization. Front Immunol 9, 1869 (2018)PubMedPubMedCentral
3.
go back to reference L. Chiossone, P.Y. Dumas, M. Vienne, E. Vivier, Natural killer cells and other innate lymphoid cells in cancer. Nat Rev Immunol 18, 671–688 (2018)PubMed L. Chiossone, P.Y. Dumas, M. Vienne, E. Vivier, Natural killer cells and other innate lymphoid cells in cancer. Nat Rev Immunol 18, 671–688 (2018)PubMed
4.
go back to reference F. Souza-Fonseca-Guimaraes, J. Cursons, N.D. Huntington, The emergence of natural killer cells as a major target in Cancer immunotherapy. Trends Immunol 40, 142–158 (2019)PubMed F. Souza-Fonseca-Guimaraes, J. Cursons, N.D. Huntington, The emergence of natural killer cells as a major target in Cancer immunotherapy. Trends Immunol 40, 142–158 (2019)PubMed
5.
go back to reference S. Sivori, P. Vacca, G. Del Zotto, E. Munari, M.C. Mingari, L. Moretta, Human NK cells: Surface receptors, inhibitory checkpoints, and translational applications. Cell Mol Immunol 16, 430–441 (2019) S. Sivori, P. Vacca, G. Del Zotto, E. Munari, M.C. Mingari, L. Moretta, Human NK cells: Surface receptors, inhibitory checkpoints, and translational applications. Cell Mol Immunol 16, 430–441 (2019)
6.
go back to reference E. Vivier, E. Tomasello, M. Baratin, T. Walzer, S. Ugolini, Functions of natural killer cells. Nat Immunol 9, 503–510 (2008)PubMed E. Vivier, E. Tomasello, M. Baratin, T. Walzer, S. Ugolini, Functions of natural killer cells. Nat Immunol 9, 503–510 (2008)PubMed
7.
go back to reference M.A. Cooper, T.A. Fehniger, M.A. Caligiuri, The biology of human natural killer-cell subsets. Trends Immunol 22, 633–640 (2001)PubMed M.A. Cooper, T.A. Fehniger, M.A. Caligiuri, The biology of human natural killer-cell subsets. Trends Immunol 22, 633–640 (2001)PubMed
8.
go back to reference M.A. Caligiuri, A. Zmuidzinas, T.J. Manley, H. Levine, K.A. Smith, J. Ritz, Functional consequences of interleukin 2 receptor expression on resting human lymphocytes. Identification of a novel natural killer cell subset with high affinity receptors J Exp Med 171, 1509–1526 (1990)PubMed M.A. Caligiuri, A. Zmuidzinas, T.J. Manley, H. Levine, K.A. Smith, J. Ritz, Functional consequences of interleukin 2 receptor expression on resting human lymphocytes. Identification of a novel natural killer cell subset with high affinity receptors J Exp Med 171, 1509–1526 (1990)PubMed
9.
go back to reference J.J. Campbell, S. Qin, D. Unutmaz, D. Soler, K.E. Murphy, M.R. Hodge, L. Wu, E.C. Butcher, Unique subpopulations of CD56+ NK and NK-T peripheral blood lymphocytes identified by chemokine receptor expression repertoire. J Immunol 166, 6477–6482 (2001)PubMed J.J. Campbell, S. Qin, D. Unutmaz, D. Soler, K.E. Murphy, M.R. Hodge, L. Wu, E.C. Butcher, Unique subpopulations of CD56+ NK and NK-T peripheral blood lymphocytes identified by chemokine receptor expression repertoire. J Immunol 166, 6477–6482 (2001)PubMed
10.
go back to reference W. Wang, A.K. Erbe, J.A. Hank, Z.S. Morris, P.M. Sondel, NK cell-mediated antibody-dependent cellular cytotoxicity in Cancer immunotherapy. Front Immunol 6, 368 (2015)PubMedPubMedCentral W. Wang, A.K. Erbe, J.A. Hank, Z.S. Morris, P.M. Sondel, NK cell-mediated antibody-dependent cellular cytotoxicity in Cancer immunotherapy. Front Immunol 6, 368 (2015)PubMedPubMedCentral
11.
go back to reference Y. Zhu, B. Huang, J. Shi, Fas ligand and lytic granule differentially control cytotoxic dynamics of natural killer cell against cancer target. Oncotarget. 7, 47163–47172 (2016)PubMedPubMedCentral Y. Zhu, B. Huang, J. Shi, Fas ligand and lytic granule differentially control cytotoxic dynamics of natural killer cell against cancer target. Oncotarget. 7, 47163–47172 (2016)PubMedPubMedCentral
12.
go back to reference E. Vivier, D.H. Raulet, A. Moretta, M.A. Caligiuri, L. Zitvogel, L.L. Lanier, W.M. Yokoyama, S. Ugolini, Innate or adaptive immunity? The example of natural killer cells Science 331, 44–49 (2011)PubMed E. Vivier, D.H. Raulet, A. Moretta, M.A. Caligiuri, L. Zitvogel, L.L. Lanier, W.M. Yokoyama, S. Ugolini, Innate or adaptive immunity? The example of natural killer cells Science 331, 44–49 (2011)PubMed
14.
go back to reference S. Lorenzo-Herrero, A. López-Soto, C. Sordo-Bahamonde, A.P. Gonzalez-Rodriguez, M. Vitale, S. Gonzalez, NK cell-based immunotherapy in Cancer metastasis. Cancers 11, 29 (2018)PubMedCentral S. Lorenzo-Herrero, A. López-Soto, C. Sordo-Bahamonde, A.P. Gonzalez-Rodriguez, M. Vitale, S. Gonzalez, NK cell-based immunotherapy in Cancer metastasis. Cancers 11, 29 (2018)PubMedCentral
15.
go back to reference Y. Yu, J. Cui, Present and future of cancer immunotherapy: A tumor microenvironmental perspective. Oncol Lett 16, 4105–4113 (2018)PubMedPubMedCentral Y. Yu, J. Cui, Present and future of cancer immunotherapy: A tumor microenvironmental perspective. Oncol Lett 16, 4105–4113 (2018)PubMedPubMedCentral
16.
go back to reference M. Wang, J. Zhao, L. Zhang, F. Wei, Y. Lian, Y. Wu, Z. Gong, S. Zhang, J. Zhou, K. Cao, X. Li, W. Xiong, G. Li, Z. Zeng, C. Guo, Role of tumor microenvironment in tumorigenesis. J Cancer 8, 761–773 (2017)PubMedPubMedCentral M. Wang, J. Zhao, L. Zhang, F. Wei, Y. Lian, Y. Wu, Z. Gong, S. Zhang, J. Zhou, K. Cao, X. Li, W. Xiong, G. Li, Z. Zeng, C. Guo, Role of tumor microenvironment in tumorigenesis. J Cancer 8, 761–773 (2017)PubMedPubMedCentral
17.
go back to reference M.B. Schaaf, A.D. Garg, P. Agostinis, Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis 9, 115 (2018)PubMedPubMedCentral M.B. Schaaf, A.D. Garg, P. Agostinis, Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis 9, 115 (2018)PubMedPubMedCentral
18.
go back to reference S. Damgaci, A. Ibrahim-Hashim, P.M. Enriquez-Navas, S. Pilon-Thomas, A. Guvenis, R.J. Gillies, Hypoxia and acidosis: Immune suppressors and therapeutic targets. Immunology. 154, 354–362 (2018)PubMedPubMedCentral S. Damgaci, A. Ibrahim-Hashim, P.M. Enriquez-Navas, S. Pilon-Thomas, A. Guvenis, R.J. Gillies, Hypoxia and acidosis: Immune suppressors and therapeutic targets. Immunology. 154, 354–362 (2018)PubMedPubMedCentral
19.
go back to reference J.D. Martin, D. Fukumura, D.G. Duda, Y. Boucher, R.K. Jain, Reengineering the tumor microenvironment to alleviate hypoxia and overcome Cancer heterogeneity. Cold Spring Harb Perspect Med 6, a027094 (2016)PubMedPubMedCentral J.D. Martin, D. Fukumura, D.G. Duda, Y. Boucher, R.K. Jain, Reengineering the tumor microenvironment to alleviate hypoxia and overcome Cancer heterogeneity. Cold Spring Harb Perspect Med 6, a027094 (2016)PubMedPubMedCentral
20.
go back to reference S.L. Shiao, A.P. Ganesan, H.S. Rugo, L.M. Coussens, Immune microenvironments in solid tumors: New targets for therapy. Genes Dev 25, 2559–2572 (2011)PubMedPubMedCentral S.L. Shiao, A.P. Ganesan, H.S. Rugo, L.M. Coussens, Immune microenvironments in solid tumors: New targets for therapy. Genes Dev 25, 2559–2572 (2011)PubMedPubMedCentral
21.
go back to reference G. Nayyar, Y. Chu, M.S. Cairo, Overcoming resistance to natural killer cell based immunotherapies for solid tumors. Front Oncol 9, 51 (2019)PubMedPubMedCentral G. Nayyar, Y. Chu, M.S. Cairo, Overcoming resistance to natural killer cell based immunotherapies for solid tumors. Front Oncol 9, 51 (2019)PubMedPubMedCentral
22.
go back to reference J. L. da Silva, A. L. S. Dos Santos, N. C. C. Nunes, F. de Moraes Lino da Silva, C. G. M. Ferreira, A. C. de Melo, Cancer immunotherapy: the art of targeting the tumor immune microenvironment. Cancer Chemother Pharmacol 84, 227–240 (2019) J. L. da Silva, A. L. S. Dos Santos, N. C. C. Nunes, F. de Moraes Lino da Silva, C. G. M. Ferreira, A. C. de Melo, Cancer immunotherapy: the art of targeting the tumor immune microenvironment. Cancer Chemother Pharmacol 84, 227–240 (2019)
23.
go back to reference S.E. Keating, V. Zaiatz-Bittencourt, R.M. Loftus, C. Keane, K. Brennan, D.K. Finlay, C.M. Gardiner, Metabolic reprogramming supports IFN-γ production by CD56bright NK cells. J Immunol 196, 2552–2560 (2016)PubMed S.E. Keating, V. Zaiatz-Bittencourt, R.M. Loftus, C. Keane, K. Brennan, D.K. Finlay, C.M. Gardiner, Metabolic reprogramming supports IFN-γ production by CD56bright NK cells. J Immunol 196, 2552–2560 (2016)PubMed
24.
go back to reference R.P. Donnelly, R.M. Loftus, S.E. Keating, K.T. Liou, C.A. Biron, C.M. Gardiner, D.K. Finlay, mTORC1-dependent metabolic reprogramming is a prerequisite for NK cell effector function. J Immunol 193, 4477–4484 (2014)PubMedPubMedCentral R.P. Donnelly, R.M. Loftus, S.E. Keating, K.T. Liou, C.A. Biron, C.M. Gardiner, D.K. Finlay, mTORC1-dependent metabolic reprogramming is a prerequisite for NK cell effector function. J Immunol 193, 4477–4484 (2014)PubMedPubMedCentral
25.
go back to reference S.M. Almutairi, A.K. Ali, W. He, D.S. Yang, P. Ghorbani, L. Wang, M.D. Fullerton, S.H. Lee, Interleukin-18 up-regulates amino acid transporters and facilitates amino acid-induced mTORC1 activation in natural killer cells. J Biol Chem 294, 4644–4655 (2019)PubMedPubMedCentral S.M. Almutairi, A.K. Ali, W. He, D.S. Yang, P. Ghorbani, L. Wang, M.D. Fullerton, S.H. Lee, Interleukin-18 up-regulates amino acid transporters and facilitates amino acid-induced mTORC1 activation in natural killer cells. J Biol Chem 294, 4644–4655 (2019)PubMedPubMedCentral
26.
go back to reference Y. Mao, V. van Hoef, X. Zhang, E. Wennerberg, J. Lorent, K. Witt, L. Masvidal, S. Liang, S. Murray, O. Larsson, R. Kiessling, A. Lundqvist, IL-15 activates mTOR and primes stress-activated gene expression leading to prolonged antitumor capacity of NK cells. Blood 128, 1475–1489 (2016) Y. Mao, V. van Hoef, X. Zhang, E. Wennerberg, J. Lorent, K. Witt, L. Masvidal, S. Liang, S. Murray, O. Larsson, R. Kiessling, A. Lundqvist, IL-15 activates mTOR and primes stress-activated gene expression leading to prolonged antitumor capacity of NK cells. Blood 128, 1475–1489 (2016)
27.
go back to reference A. Marçais, J. Cherfils-Vicini, C. Viant, S. Degouve, S. Viel, A. Fenis, J. Rabilloud, K. Mayol, A. Tavares, J. Bienvenu, Y.G. Gangloff, E. Gilson, E. Vivier, T. Walzer, The metabolic checkpoint kinase mTOR is essential for IL-15 signaling during the development and activation of NK cells. Nat Immunol 15, 749–757 (2014)PubMedPubMedCentral A. Marçais, J. Cherfils-Vicini, C. Viant, S. Degouve, S. Viel, A. Fenis, J. Rabilloud, K. Mayol, A. Tavares, J. Bienvenu, Y.G. Gangloff, E. Gilson, E. Vivier, T. Walzer, The metabolic checkpoint kinase mTOR is essential for IL-15 signaling during the development and activation of NK cells. Nat Immunol 15, 749–757 (2014)PubMedPubMedCentral
28.
go back to reference Y. Wu, Z. Tian, H. Wei, Developmental and functional control of natural killer cells by cytokines. Front Immunol 8, 930 (2017)PubMedPubMedCentral Y. Wu, Z. Tian, H. Wei, Developmental and functional control of natural killer cells by cytokines. Front Immunol 8, 930 (2017)PubMedPubMedCentral
29.
go back to reference N.W. Zwirner, A. Ziblat, Regulation of NK cell activation and effector functions by the IL-12 family of cytokines: The case of IL-27. Front Immunol 8, 25 (2017)PubMedPubMedCentral N.W. Zwirner, A. Ziblat, Regulation of NK cell activation and effector functions by the IL-12 family of cytokines: The case of IL-27. Front Immunol 8, 25 (2017)PubMedPubMedCentral
30.
go back to reference J. Rautela, N.D. Huntington, IL-15 signaling in NK cell cancer immunotherapy. Curr Opin Immunol 44, 1–6 (2017)PubMed J. Rautela, N.D. Huntington, IL-15 signaling in NK cell cancer immunotherapy. Curr Opin Immunol 44, 1–6 (2017)PubMed
31.
go back to reference M. Patidar, N. Yadav, S.K. Dalai, Interleukin 15: A key cytokine for immunotherapy. Cytokine Growth Factor Rev 31, 49–59 (2016)PubMed M. Patidar, N. Yadav, S.K. Dalai, Interleukin 15: A key cytokine for immunotherapy. Cytokine Growth Factor Rev 31, 49–59 (2016)PubMed
32.
go back to reference N. Nandagopal, A.K. Ali, A.K. Komal, S.H. Lee, The critical role of IL-15–PI3K–mTOR pathway in natural killer cell effector functions. Front Immunol 5, 187 (2014)PubMedPubMedCentral N. Nandagopal, A.K. Ali, A.K. Komal, S.H. Lee, The critical role of IL-15–PI3K–mTOR pathway in natural killer cell effector functions. Front Immunol 5, 187 (2014)PubMedPubMedCentral
33.
go back to reference A.K. Ali, N. Nandagopal, S.H. Lee, IL-15–PI3K–AKT–mTOR: A critical pathway in the life journey of natural killer cells. Front Immunol 6, 355 (2015)PubMedPubMedCentral A.K. Ali, N. Nandagopal, S.H. Lee, IL-15–PI3K–AKT–mTOR: A critical pathway in the life journey of natural killer cells. Front Immunol 6, 355 (2015)PubMedPubMedCentral
34.
go back to reference R. Sharma, A. Das, IL-2 mediates NK cell proliferation but not hyperactivity. Immunol Res 66, 151–157 (2018)PubMed R. Sharma, A. Das, IL-2 mediates NK cell proliferation but not hyperactivity. Immunol Res 66, 151–157 (2018)PubMed
35.
go back to reference A.C. Jaime-Ramirez, B.L. Mundy-Bosse, S. Kondadasula, N.B. Jones, J.M. Roda, A. Mani, R. Parihar, V. Karpa, T.L. Papenfuss, K.M. LaPerle, E. Biller, A. Lehman, A.R. Chaudhury, D. Jarjoura, R.W. Burry, W.E. Carson, IL-12 enhances the antitumor actions of Trastuzumab via NK cell IFN-γ production. J Immunol 186, 3401–3409 (2011)PubMedPubMedCentral A.C. Jaime-Ramirez, B.L. Mundy-Bosse, S. Kondadasula, N.B. Jones, J.M. Roda, A. Mani, R. Parihar, V. Karpa, T.L. Papenfuss, K.M. LaPerle, E. Biller, A. Lehman, A.R. Chaudhury, D. Jarjoura, R.W. Burry, W.E. Carson, IL-12 enhances the antitumor actions of Trastuzumab via NK cell IFN-γ production. J Immunol 186, 3401–3409 (2011)PubMedPubMedCentral
36.
go back to reference Y.H. Choi, E.J. Lim, S.W. Kim, Y.W. Moon, K.S. Park, H.J. An, IL-27 enhances IL-15/IL-18-mediated activation of human natural killer cells. J. ImmunoTher. Cancer 7, 168 (2019)PubMedPubMedCentral Y.H. Choi, E.J. Lim, S.W. Kim, Y.W. Moon, K.S. Park, H.J. An, IL-27 enhances IL-15/IL-18-mediated activation of human natural killer cells. J. ImmunoTher. Cancer 7, 168 (2019)PubMedPubMedCentral
37.
go back to reference A. Ziblat, C.I. Domaica, R.G. Spallanzani, X.L.R. Iraolagoitia, L.E. Rossi, D.E. Avila, N.I. Torres, M.B. Fuertes, N.W. Zwirner, IL-27 stimulates human NK-cell effector functions and primes NK cells for IL-18 responsiveness. Eur J Immunol 45, 192–202 (2015)PubMed A. Ziblat, C.I. Domaica, R.G. Spallanzani, X.L.R. Iraolagoitia, L.E. Rossi, D.E. Avila, N.I. Torres, M.B. Fuertes, N.W. Zwirner, IL-27 stimulates human NK-cell effector functions and primes NK cells for IL-18 responsiveness. Eur J Immunol 45, 192–202 (2015)PubMed
38.
go back to reference N.W. Zwirner, C.I. Domaica, Cytokine regulation of natural killer cell effector functions. Biofactors 36, 274–288 (2010) N.W. Zwirner, C.I. Domaica, Cytokine regulation of natural killer cell effector functions. Biofactors 36, 274–288 (2010)
39.
go back to reference M.P. Keppel, N. Topcagic, A.Y. Mah, T.P. Vogel, M.A. Cooper, Activation-specific metabolic requirements for NK cell IFN-γ production. J Immunol 194, 1954–1962 (2015)PubMedPubMedCentral M.P. Keppel, N. Topcagic, A.Y. Mah, T.P. Vogel, M.A. Cooper, Activation-specific metabolic requirements for NK cell IFN-γ production. J Immunol 194, 1954–1962 (2015)PubMedPubMedCentral
40.
go back to reference A.Y. Mah, M.A. Cooper, Metabolic regulation of natural killer cell IFN-γ production. Crit Rev Immunol 36, 131–147 (2016)PubMedPubMedCentral A.Y. Mah, M.A. Cooper, Metabolic regulation of natural killer cell IFN-γ production. Crit Rev Immunol 36, 131–147 (2016)PubMedPubMedCentral
41.
go back to reference A.M. Chambers, K.B. Lupo, S. Matosevic, Tumor microenvironment-induced Immunometabolic reprogramming of natural killer cells. Front Immunol 9, 2517 (2018)PubMedPubMedCentral A.M. Chambers, K.B. Lupo, S. Matosevic, Tumor microenvironment-induced Immunometabolic reprogramming of natural killer cells. Front Immunol 9, 2517 (2018)PubMedPubMedCentral
42.
go back to reference W. Salzberger, G. Martrus, K. Bachmann, H. Goebels, L. Heß, M. Koch, A. Langeneckert, S. Lunemann, K.J. Oldhafer, C. Pfeifer, T. Poch, L. Richert, C. Schramm, R. Wahib, M.J. Bunders, M. Altfeld, Tissue-resident NK cells differ in their expression profile of the nutrient transporters Glut1, CD98 and CD71. PLoS One 13, e0201170 (2018)PubMedPubMedCentral W. Salzberger, G. Martrus, K. Bachmann, H. Goebels, L. Heß, M. Koch, A. Langeneckert, S. Lunemann, K.J. Oldhafer, C. Pfeifer, T. Poch, L. Richert, C. Schramm, R. Wahib, M.J. Bunders, M. Altfeld, Tissue-resident NK cells differ in their expression profile of the nutrient transporters Glut1, CD98 and CD71. PLoS One 13, e0201170 (2018)PubMedPubMedCentral
43.
go back to reference J. Cong, X. Wang, X. Zheng, D. Wang, B. Fu, R. Sun, Z. Tian, H. Wei, Dysfunction of natural killer cells by FBP1-induced inhibition of glycolysis during lung Cancer progression. Cell Metab 28, 243–255 (2018)PubMed J. Cong, X. Wang, X. Zheng, D. Wang, B. Fu, R. Sun, Z. Tian, H. Wei, Dysfunction of natural killer cells by FBP1-induced inhibition of glycolysis during lung Cancer progression. Cell Metab 28, 243–255 (2018)PubMed
44.
go back to reference K. Slattery, V. Zaiatz-Bittencourt, E. Woods, K. Brennan, S. Marks, S. Chew, M. Conroy, C. Goggin, J. Kennedy, D.K. Finlay, C.M. Gardiner, TGFβ drives mitochondrial dysfunction in peripheral blood NK cells during metastatic breast cancer. BioRxiv 648501 (2019) K. Slattery, V. Zaiatz-Bittencourt, E. Woods, K. Brennan, S. Marks, S. Chew, M. Conroy, C. Goggin, J. Kennedy, D.K. Finlay, C.M. Gardiner, TGFβ drives mitochondrial dysfunction in peripheral blood NK cells during metastatic breast cancer. BioRxiv 648501 (2019)
45.
go back to reference A.M. Chambers, J. Wang, K.B. Lupo, H. Yu, N.M. Atallah Lanman, S. Matosevic, Adenosinergic signaling alters natural killer cell functional responses. Front Immunol 9, 2533 (2018)PubMedPubMedCentral A.M. Chambers, J. Wang, K.B. Lupo, H. Yu, N.M. Atallah Lanman, S. Matosevic, Adenosinergic signaling alters natural killer cell functional responses. Front Immunol 9, 2533 (2018)PubMedPubMedCentral
46.
go back to reference S. Matosevic, A.M. Chambers, Immunometabolic dysfunction of natural killer cells mediated by the hypoxia-CD73 axis in solid tumors. Front Mol Biosci 6, 60 (2019)PubMedPubMedCentral S. Matosevic, A.M. Chambers, Immunometabolic dysfunction of natural killer cells mediated by the hypoxia-CD73 axis in solid tumors. Front Mol Biosci 6, 60 (2019)PubMedPubMedCentral
47.
go back to reference M. Balsamo, C. Manzini, G. Pietra, F. Raggi, F. Blengio, M.C. Mingari, L. Varesio, L. Moretta, M.C. Bosco, M. Vitale, Hypoxia downregulates the expression of activating receptors involved in NK-cell-mediated target cell killing without affecting ADCC. Eur J Immunol 43, 2756–2764 (2013)PubMed M. Balsamo, C. Manzini, G. Pietra, F. Raggi, F. Blengio, M.C. Mingari, L. Varesio, L. Moretta, M.C. Bosco, M. Vitale, Hypoxia downregulates the expression of activating receptors involved in NK-cell-mediated target cell killing without affecting ADCC. Eur J Immunol 43, 2756–2764 (2013)PubMed
48.
go back to reference S.Y. Velásquez, D. Killian, J. Schulte, C. Sticht, M. Thiel, H.A. Lindner, Short-term hypoxia synergizes with interleukin 15 priming in driving glycolytic gene transcription and supports human natural killer cell activities. J Biol Chem 291, 12960–12977 (2016)PubMedPubMedCentral S.Y. Velásquez, D. Killian, J. Schulte, C. Sticht, M. Thiel, H.A. Lindner, Short-term hypoxia synergizes with interleukin 15 priming in driving glycolytic gene transcription and supports human natural killer cell activities. J Biol Chem 291, 12960–12977 (2016)PubMedPubMedCentral
49.
go back to reference E. Krzywinska, C. Kantari-Mimoun, Y. Kerdiles, M. Sobecki, T. Isagawa, D. Gotthardt, M. Castells, J. Haubold, C. Millien, T. Viel, B. Tavitian, N. Takeda, J. Fandrey, E. Vivier, V. Sexl, C. Stockmann, Loss of HIF-1α in natural killer cells inhibits tumour growth by stimulating non-productive angiogenesis. Nat Commun 8, 1597 (2017)PubMedPubMedCentral E. Krzywinska, C. Kantari-Mimoun, Y. Kerdiles, M. Sobecki, T. Isagawa, D. Gotthardt, M. Castells, J. Haubold, C. Millien, T. Viel, B. Tavitian, N. Takeda, J. Fandrey, E. Vivier, V. Sexl, C. Stockmann, Loss of HIF-1α in natural killer cells inhibits tumour growth by stimulating non-productive angiogenesis. Nat Commun 8, 1597 (2017)PubMedPubMedCentral
50.
go back to reference Y. Feng, Y. Xiong, T. Qiao, X. Li, L. Jia, Y. Han, Lactate dehydrogenase a: A key player in carcinogenesis and potential target in cancer therapy. Cancer Med 7, 6124–6136 (2018)PubMedPubMedCentral Y. Feng, Y. Xiong, T. Qiao, X. Li, L. Jia, Y. Han, Lactate dehydrogenase a: A key player in carcinogenesis and potential target in cancer therapy. Cancer Med 7, 6124–6136 (2018)PubMedPubMedCentral
51.
go back to reference C. Harmon, M.W. Robinson, F. Hand, D. Almuaili, K. Mentor, D.D. Houlihan, E. Hoti, L. Lynch, J. Geoghegan, C. O’Farrelly, Lactate-mediated acidification of tumor microenvironment induces apoptosis of liver-resident NK cells in colorectal liver metastasis. Cancer Immunol Res. 7, 335–346 (2019)PubMed C. Harmon, M.W. Robinson, F. Hand, D. Almuaili, K. Mentor, D.D. Houlihan, E. Hoti, L. Lynch, J. Geoghegan, C. O’Farrelly, Lactate-mediated acidification of tumor microenvironment induces apoptosis of liver-resident NK cells in colorectal liver metastasis. Cancer Immunol Res. 7, 335–346 (2019)PubMed
52.
go back to reference J. Wang, S. Matosevic, NT5E/CD73 as correlative factor of patient survival and natural killer cell infiltration in glioblastoma. J Clin Med 8, 1526 (2019)PubMedCentral J. Wang, S. Matosevic, NT5E/CD73 as correlative factor of patient survival and natural killer cell infiltration in glioblastoma. J Clin Med 8, 1526 (2019)PubMedCentral
53.
go back to reference J.S. Schleypen, N. Baur, R. Kammerer, P.J. Nelson, K. Rohrmann, E.F. Gröne, M. Hohenfellner, A. Haferkamp, H. Pohla, D.J. Schendel, C.S. Falk, E. Noessner, Cytotoxic markers and frequency predict functional capacity of natural killer cells infiltrating renal cell carcinoma. Clin Cancer Res 12, 718–725 (2006)PubMed J.S. Schleypen, N. Baur, R. Kammerer, P.J. Nelson, K. Rohrmann, E.F. Gröne, M. Hohenfellner, A. Haferkamp, H. Pohla, D.J. Schendel, C.S. Falk, E. Noessner, Cytotoxic markers and frequency predict functional capacity of natural killer cells infiltrating renal cell carcinoma. Clin Cancer Res 12, 718–725 (2006)PubMed
54.
go back to reference J.S. Schleypen, M. Von Geldern, E.H. Weiss, N. Kotzias, K. Rohrmann, D.J. Schendel, C.S. Falk, H. Pohla, Renal cell carcinoma-infiltrating natural killer cells express differential repertoires of activating and inhibitory receptors and are inhibited by specific HLA class I allotypes. Int J Cancer 106, 905–912 (2003)PubMed J.S. Schleypen, M. Von Geldern, E.H. Weiss, N. Kotzias, K. Rohrmann, D.J. Schendel, C.S. Falk, H. Pohla, Renal cell carcinoma-infiltrating natural killer cells express differential repertoires of activating and inhibitory receptors and are inhibited by specific HLA class I allotypes. Int J Cancer 106, 905–912 (2003)PubMed
55.
go back to reference P.I. Tartter, B. Steinberg, D.M. Barron, G. Martinelli, The prognostic significance of natural killer cytotoxicity in patients with colorectal cancer. Arch Surg 122, 1264–1268 (1987)PubMed P.I. Tartter, B. Steinberg, D.M. Barron, G. Martinelli, The prognostic significance of natural killer cytotoxicity in patients with colorectal cancer. Arch Surg 122, 1264–1268 (1987)PubMed
56.
go back to reference C. Denkert, G. von Minckwitz, S. Darb-Esfahani, B. Lederer, B. I. Heppner, K. E. Weber, J. Budczies, J. Huober, F. Klauschen, J. Furlanetto, W. D. Schmitt, J. U. Blohmer, T. Karn, B. M. Pfitzner, S. Kümmel, K. Engels, A. Schneeweiss, A. Hartmann, A. Noske, P. A. Fasching, C. Jackisch, M. van Mackelenbergh, P. Sinn, C. Schem, C. Hanusch, M. Untch, S. Loibl, Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol 19, 40–50 (2018) C. Denkert, G. von Minckwitz, S. Darb-Esfahani, B. Lederer, B. I. Heppner, K. E. Weber, J. Budczies, J. Huober, F. Klauschen, J. Furlanetto, W. D. Schmitt, J. U. Blohmer, T. Karn, B. M. Pfitzner, S. Kümmel, K. Engels, A. Schneeweiss, A. Hartmann, A. Noske, P. A. Fasching, C. Jackisch, M. van Mackelenbergh, P. Sinn, C. Schem, C. Hanusch, M. Untch, S. Loibl, Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol 19, 40–50 (2018)
57.
go back to reference R. Salgado, C. Denkert, C. Campbell, P. Savas, P. Nuciforo, P. Nucifero, C. Aura, E. de Azambuja, H. Eidtmann, C.E. Ellis, J. Baselga, M.J. Piccart-Gebhart, S. Michiels, I. Bradbury, C. Sotiriou, S. Loi, Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast Cancer treated with Lapatinib and Trastuzumab: A secondary analysis of the NeoALTTO trial. JAMA Oncol 1, 448–454 (2015)PubMedPubMedCentral R. Salgado, C. Denkert, C. Campbell, P. Savas, P. Nuciforo, P. Nucifero, C. Aura, E. de Azambuja, H. Eidtmann, C.E. Ellis, J. Baselga, M.J. Piccart-Gebhart, S. Michiels, I. Bradbury, C. Sotiriou, S. Loi, Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast Cancer treated with Lapatinib and Trastuzumab: A secondary analysis of the NeoALTTO trial. JAMA Oncol 1, 448–454 (2015)PubMedPubMedCentral
58.
go back to reference R. Gennari, S. Menard, F. Fagnoni, L. Ponchio, M. Scelsi, E. Tagliabue, F. Castiglioni, L. Villani, C. Magalotti, N. Gibelli, B. Oliviero, B. Ballardini, G. Da Prada, A. Zambelli, A. Costa, Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res 10, 5650–5655 (2004)PubMed R. Gennari, S. Menard, F. Fagnoni, L. Ponchio, M. Scelsi, E. Tagliabue, F. Castiglioni, L. Villani, C. Magalotti, N. Gibelli, B. Oliviero, B. Ballardini, G. Da Prada, A. Zambelli, A. Costa, Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res 10, 5650–5655 (2004)PubMed
59.
go back to reference A. Muntasell, F. Rojo, S. Servitja, C. Rubio-Perez, M. Cabo, D. Tamborero, M. Costa-García, M. Martínez-Garcia, S. Menéndez, I. Vazquez, A. Lluch, A. Gonzalez-Perez, A. Rovira, M. López-Botet, J. Albanell, NK cell infiltrates and HLA class I expression in primary HER2+ breast Cancer predict and uncouple pathological response and disease-free survival. Clin Cancer Res 25, 1535–1545 (2019)PubMed A. Muntasell, F. Rojo, S. Servitja, C. Rubio-Perez, M. Cabo, D. Tamborero, M. Costa-García, M. Martínez-Garcia, S. Menéndez, I. Vazquez, A. Lluch, A. Gonzalez-Perez, A. Rovira, M. López-Botet, J. Albanell, NK cell infiltrates and HLA class I expression in primary HER2+ breast Cancer predict and uncouple pathological response and disease-free survival. Clin Cancer Res 25, 1535–1545 (2019)PubMed
60.
go back to reference J. Cursons, F. Souza-Fonseca-Guimaraes, M. Foroutan, A. Anderson, F. Hollande, S. Hediyeh-Zadeh, A. Behren, N.D. Huntington, M.J. Davis, A gene signature predicting natural killer cell infiltration and improved survival in melanoma patients. Cancer Immunol Res 7, 1162–1174 (2019)PubMed J. Cursons, F. Souza-Fonseca-Guimaraes, M. Foroutan, A. Anderson, F. Hollande, S. Hediyeh-Zadeh, A. Behren, N.D. Huntington, M.J. Davis, A gene signature predicting natural killer cell infiltration and improved survival in melanoma patients. Cancer Immunol Res 7, 1162–1174 (2019)PubMed
61.
go back to reference A. Gras Navarro, A.T. Björklund, M. Chekenya, Therapeutic potential and challenges of natural killer cells in treatment of solid tumors. Front Immunol 6, 102 (2015) A. Gras Navarro, A.T. Björklund, M. Chekenya, Therapeutic potential and challenges of natural killer cells in treatment of solid tumors. Front Immunol 6, 102 (2015)
62.
go back to reference A. Jiménez-Sánchez, D. Memon, S. Pourpe, H. Veeraraghavan, Y. Li, H.A. Vargas, M.B. Gill, K.J. Park, O. Zivanovic, J. Konner, J. Ricca, D. Zamarin, T. Walther, C. Aghajanian, J.D. Wolchok, E. Sala, T. Merghoub, A. Snyder, M.L. Miller, Heterogeneous tumor-immune microenvironments among differentially growing metastases in an ovarian Cancer patient. Cell 170, 927–938 (2017) A. Jiménez-Sánchez, D. Memon, S. Pourpe, H. Veeraraghavan, Y. Li, H.A. Vargas, M.B. Gill, K.J. Park, O. Zivanovic, J. Konner, J. Ricca, D. Zamarin, T. Walther, C. Aghajanian, J.D. Wolchok, E. Sala, T. Merghoub, A. Snyder, M.L. Miller, Heterogeneous tumor-immune microenvironments among differentially growing metastases in an ovarian Cancer patient. Cell 170, 927–938 (2017)
63.
go back to reference J. Kmiecik, J. Zimmer, M. Chekenya, Natural killer cells in intracranial neoplasms: Presence and therapeutic efficacy against brain tumours. J Neuro-Oncol 116, 1–9 (2014) J. Kmiecik, J. Zimmer, M. Chekenya, Natural killer cells in intracranial neoplasms: Presence and therapeutic efficacy against brain tumours. J Neuro-Oncol 116, 1–9 (2014)
64.
go back to reference I. Golán, L. Rodríguez de la Fuente, J.A. Costoya, NK Cell-Based Glioblastoma Immunotherapy Cancers 10, 522 (2018) I. Golán, L. Rodríguez de la Fuente, J.A. Costoya, NK Cell-Based Glioblastoma Immunotherapy Cancers 10, 522 (2018)
65.
go back to reference W. Tomaszewski, L. Sanchez-Perez, T.F. Gajewski, J.H. Sampson, Brain tumor microenvironment and host state: Implications for immunotherapy. Clin Cancer Res 25, 4202–4210 (2019)PubMed W. Tomaszewski, L. Sanchez-Perez, T.F. Gajewski, J.H. Sampson, Brain tumor microenvironment and host state: Implications for immunotherapy. Clin Cancer Res 25, 4202–4210 (2019)PubMed
66.
go back to reference R. Castriconi, A. Dondero, F. Bellora, L. Moretta, A. Castellano, F. Locatelli, M.V. Corrias, A. Moretta, C. Bottino, Neuroblastoma-derived TGF-β1 modulates the chemokine receptor repertoire of human resting NK cells. J Immunol 190, 5321–5328 (2013)PubMed R. Castriconi, A. Dondero, F. Bellora, L. Moretta, A. Castellano, F. Locatelli, M.V. Corrias, A. Moretta, C. Bottino, Neuroblastoma-derived TGF-β1 modulates the chemokine receptor repertoire of human resting NK cells. J Immunol 190, 5321–5328 (2013)PubMed
67.
go back to reference I. Melero, A. Rouzaut, G.T. Motz, G. Coukos, T-cell and NK-cell infiltration into solid tumors: A key limiting factor for efficacious cancer immunotherapy. Cancer Discov 4, 522–526 (2014) I. Melero, A. Rouzaut, G.T. Motz, G. Coukos, T-cell and NK-cell infiltration into solid tumors: A key limiting factor for efficacious cancer immunotherapy. Cancer Discov 4, 522–526 (2014)
68.
go back to reference J. Eckl, A. Buchner, P.U. Prinz, R. Riesenberg, S.I. Siegert, R. Kammerer, P.J. Nelson, E. Noessner, Transcript signature predicts tissue NK cell content and defines renal cell carcinoma subgroups independent of TNM staging. J Mol Med 90, 55–66 (2012)PubMed J. Eckl, A. Buchner, P.U. Prinz, R. Riesenberg, S.I. Siegert, R. Kammerer, P.J. Nelson, E. Noessner, Transcript signature predicts tissue NK cell content and defines renal cell carcinoma subgroups independent of TNM staging. J Mol Med 90, 55–66 (2012)PubMed
69.
go back to reference S.P. Schantz, N.G. Ordonez, Quantitation of natural killer cell function and risk of metastatic poorly differentiated head and neck cancer. Nat Immun Cell Growth Regul 10, 278–288 (1991)PubMed S.P. Schantz, N.G. Ordonez, Quantitation of natural killer cell function and risk of metastatic poorly differentiated head and neck cancer. Nat Immun Cell Growth Regul 10, 278–288 (1991)PubMed
70.
go back to reference P. Carrega, B. Morandi, R. Costa, G. Frumento, G. Forte, G. Altavilla, G.B. Ratto, M.C. Mingari, L. Moretta, G. Ferlazzo, Natural killer cells infiltrating human nonsmall-cell lung cancer are enriched in CD56 bright CD16(−) cells and display an impaired capability to kill tumor cells. Cancer 112, 863–875 (2008)PubMed P. Carrega, B. Morandi, R. Costa, G. Frumento, G. Forte, G. Altavilla, G.B. Ratto, M.C. Mingari, L. Moretta, G. Ferlazzo, Natural killer cells infiltrating human nonsmall-cell lung cancer are enriched in CD56 bright CD16(−) cells and display an impaired capability to kill tumor cells. Cancer 112, 863–875 (2008)PubMed
71.
go back to reference K.B. Lupo, S. Matosevic, Natural killer cells as allogeneic effectors in adoptive Cancer immunotherapy. Cancers 11, 6 (2019) K.B. Lupo, S. Matosevic, Natural killer cells as allogeneic effectors in adoptive Cancer immunotherapy. Cancers 11, 6 (2019)
72.
go back to reference A. Stojanovic, A. Cerwenka, Natural killer cells and solid tumors. J Innate Immun 3, 355–364 (2011)PubMed A. Stojanovic, A. Cerwenka, Natural killer cells and solid tumors. J Innate Immun 3, 355–364 (2011)PubMed
73.
go back to reference G. Habif, A. Crinier, P. André, E. Vivier, E. Narni-Mancinelli, Targeting natural killer cells in solid tumors. Cell Mol Immunol 16, 415–422 (2019)PubMedPubMedCentral G. Habif, A. Crinier, P. André, E. Vivier, E. Narni-Mancinelli, Targeting natural killer cells in solid tumors. Cell Mol Immunol 16, 415–422 (2019)PubMedPubMedCentral
74.
go back to reference T.N. Dao, S. Matosevic, Immunometabolic responses of natural killer cells to inhibitory tumor microenvironment checkpoints. Immunometabolism 1, e190003 (2019) T.N. Dao, S. Matosevic, Immunometabolic responses of natural killer cells to inhibitory tumor microenvironment checkpoints. Immunometabolism 1, e190003 (2019)
75.
go back to reference A. Zingoni, R. Molfetta, C. Fionda, A. Soriani, R. Paolini, M. Cippitelli, C. Cerboni, A. Santoni, NKG2D and Its Ligands: “One for All, All for One.” Front Immunol 9, 476 (2018) A. Zingoni, R. Molfetta, C. Fionda, A. Soriani, R. Paolini, M. Cippitelli, C. Cerboni, A. Santoni, NKG2D and Its Ligands: “One for All, All for One.” Front Immunol 9, 476 (2018)
76.
go back to reference C.A. Crane, S.J. Han, J.J. Barry, B.J. Ahn, L.L. Lanier, A.T. Parsa, TGF-β downregulates the activating receptor NKG2D on NK cells and CD8+ T cells in glioma patients. Neuro-Oncology 12, 7–13 (2009) C.A. Crane, S.J. Han, J.J. Barry, B.J. Ahn, L.L. Lanier, A.T. Parsa, TGF-β downregulates the activating receptor NKG2D on NK cells and CD8+ T cells in glioma patients. Neuro-Oncology 12, 7–13 (2009)
77.
go back to reference J.C. Lee, K.M. Lee, D.W. Kim, D.S. Heo, Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J Immunol 172, 7335–7340 (2004)PubMed J.C. Lee, K.M. Lee, D.W. Kim, D.S. Heo, Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J Immunol 172, 7335–7340 (2004)PubMed
78.
go back to reference C.D. Cluxton, C. Spillane, S.A. O’Toole, O. Sheils, C.M. Gardiner, J.J. O’Leary, Suppression of natural killer cell NKG2D and CD226 anti-tumour cascades by platelet cloaked cancer cells: Implications for the metastatic cascade. PLoS One 14, e0211538 (2019)PubMedPubMedCentral C.D. Cluxton, C. Spillane, S.A. O’Toole, O. Sheils, C.M. Gardiner, J.J. O’Leary, Suppression of natural killer cell NKG2D and CD226 anti-tumour cascades by platelet cloaked cancer cells: Implications for the metastatic cascade. PLoS One 14, e0211538 (2019)PubMedPubMedCentral
79.
go back to reference H. Saito, T. Osaki, M. Ikeguchi, Decreased NKG2D expression on NK cells correlates with impaired NK cell function in patients with gastric cancer. Gastric Cancer 15, 27–33 (2012)PubMed H. Saito, T. Osaki, M. Ikeguchi, Decreased NKG2D expression on NK cells correlates with impaired NK cell function in patients with gastric cancer. Gastric Cancer 15, 27–33 (2012)PubMed
80.
go back to reference D. Schmiedel, O. Mandelboim, NKG2D ligands-critical targets for Cancer immune escape and therapy. Front Immunol 9, 2040 (2018)PubMedPubMedCentral D. Schmiedel, O. Mandelboim, NKG2D ligands-critical targets for Cancer immune escape and therapy. Front Immunol 9, 2040 (2018)PubMedPubMedCentral
81.
go back to reference E.M. de Kruijf, A. Sajet, J.G.H. van Nes, H. Putter, V.T.H.B.M. Smit, R.A. Eagle, I. Jafferji, J. Trowsdale, G.J. Liefers, C.J.H. van de Velde, P.J.K. Kuppen, NKG2D ligand tumor expression and association with clinical outcome in early breast cancer patients: An observational study. BMC Cancer 12, 24 (2012) E.M. de Kruijf, A. Sajet, J.G.H. van Nes, H. Putter, V.T.H.B.M. Smit, R.A. Eagle, I. Jafferji, J. Trowsdale, G.J. Liefers, C.J.H. van de Velde, P.J.K. Kuppen, NKG2D ligand tumor expression and association with clinical outcome in early breast cancer patients: An observational study. BMC Cancer 12, 24 (2012)
82.
go back to reference G. Chitadze, J. Bhat, M. Lettau, O. Janssen, D. Kabelitz, Generation of soluble NKG2D ligands: Proteolytic cleavage, exosome secretion and functional implications. Scand J Immunol 78, 120–129 (2013)PubMed G. Chitadze, J. Bhat, M. Lettau, O. Janssen, D. Kabelitz, Generation of soluble NKG2D ligands: Proteolytic cleavage, exosome secretion and functional implications. Scand J Immunol 78, 120–129 (2013)PubMed
83.
go back to reference K. Kono, A. Takahashi, F. Ichihara, H. Sugai, H. Fujii, Y. Matsumoto, Impaired antibody-dependent cellular cytotoxicity mediated by herceptin in patients with gastric cancer. Cancer Res 62, 5813–5817 (2002)PubMed K. Kono, A. Takahashi, F. Ichihara, H. Sugai, H. Fujii, Y. Matsumoto, Impaired antibody-dependent cellular cytotoxicity mediated by herceptin in patients with gastric cancer. Cancer Res 62, 5813–5817 (2002)PubMed
84.
go back to reference G. Peruzzi, L. Femnou, A. Gil-Krzewska, F. Borrego, J. Weck, K. Krzewski, J.E. Coligan, Membrane-type 6 matrix metalloproteinase regulates the activation-induced downmodulation of CD16 in human primary NK cells. J Immunol 191, 1883–1894 (2013)PubMedPubMedCentral G. Peruzzi, L. Femnou, A. Gil-Krzewska, F. Borrego, J. Weck, K. Krzewski, J.E. Coligan, Membrane-type 6 matrix metalloproteinase regulates the activation-induced downmodulation of CD16 in human primary NK cells. J Immunol 191, 1883–1894 (2013)PubMedPubMedCentral
85.
go back to reference R. Romee, B. Foley, T. Lenvik, Y. Wang, B. Zhang, D. Ankarlo, X. Luo, S. Cooley, M. Verneris, B. Walcheck, J. Miller, NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood 121, 3599–3608 (2013) R. Romee, B. Foley, T. Lenvik, Y. Wang, B. Zhang, D. Ankarlo, X. Luo, S. Cooley, M. Verneris, B. Walcheck, J. Miller, NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood 121, 3599–3608 (2013)
86.
go back to reference P. Darvin, S.M. Toor, V. Sasidharan Nair, E. Elkord, Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp Mol Med 50, 165 (2018)PubMedCentral P. Darvin, S.M. Toor, V. Sasidharan Nair, E. Elkord, Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp Mol Med 50, 165 (2018)PubMedCentral
87.
go back to reference S.C. Wei, C.R. Duffy, J.P. Allison, Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov 8, 1069–1086 (2018) S.C. Wei, C.R. Duffy, J.P. Allison, Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov 8, 1069–1086 (2018)
88.
go back to reference T. Kamiya, S.V. Seow, D. Wong, M. Robinson, D. Campana, Blocking expression of inhibitory receptor NKG2A overcomes tumor resistance to NK cells. J Clin Invest 130, 2094–2106 (2019) T. Kamiya, S.V. Seow, D. Wong, M. Robinson, D. Campana, Blocking expression of inhibitory receptor NKG2A overcomes tumor resistance to NK cells. J Clin Invest 130, 2094–2106 (2019)
89.
go back to reference E. Le Dréan, F. Vély, L. Olcese, A. Cambiaggi, S. Guia, G. Krystal, N. Gervois, A. Moretta, F. Jotereau, E. Vivier, Inhibition of antigen-induced T cell response and antibody-induced NK cell cytotoxicity by NKG2A: Association of NKG2A with SHP-1 and SHP-2 protein-tyrosine phosphatases. Eur J Immunol 28, 264–276 (1998)PubMed E. Le Dréan, F. Vély, L. Olcese, A. Cambiaggi, S. Guia, G. Krystal, N. Gervois, A. Moretta, F. Jotereau, E. Vivier, Inhibition of antigen-induced T cell response and antibody-induced NK cell cytotoxicity by NKG2A: Association of NKG2A with SHP-1 and SHP-2 protein-tyrosine phosphatases. Eur J Immunol 28, 264–276 (1998)PubMed
90.
go back to reference F. Borrego, M. Masilamani, J. Kabat, T.B. Sanni, J.E. Coligan, The cell biology of the human natural killer cell CD94/NKG2A inhibitory receptor. Mol Immunol 42, 485–488 (2005)PubMed F. Borrego, M. Masilamani, J. Kabat, T.B. Sanni, J.E. Coligan, The cell biology of the human natural killer cell CD94/NKG2A inhibitory receptor. Mol Immunol 42, 485–488 (2005)PubMed
91.
go back to reference M. Valés-Gómez, H.T. Reyburn, R.A. Erskine, M. López-Botet, J.L. Strominger, Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-a and the activating receptor CD94/NKG2-C to HLA-E. EMBO J 18, 4250–4260 (1999)PubMedPubMedCentral M. Valés-Gómez, H.T. Reyburn, R.A. Erskine, M. López-Botet, J.L. Strominger, Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-a and the activating receptor CD94/NKG2-C to HLA-E. EMBO J 18, 4250–4260 (1999)PubMedPubMedCentral
92.
go back to reference H. Wada, N. Matsumoto, K. Maenaka, K. Suzuki, K. Yamamoto, The inhibitory NK cell receptor CD94/NKG2A and the activating receptor CD94/NKG2C bind the top of HLA-E through mostly shared but partly distinct sets of HLA-E residues. Eur J Immunol 34, 81–90 (2004)PubMed H. Wada, N. Matsumoto, K. Maenaka, K. Suzuki, K. Yamamoto, The inhibitory NK cell receptor CD94/NKG2A and the activating receptor CD94/NKG2C bind the top of HLA-E through mostly shared but partly distinct sets of HLA-E residues. Eur J Immunol 34, 81–90 (2004)PubMed
93.
go back to reference C. Sun, J. Xu, Q. Huang, M. Huang, H. Wen, C. Zhang, J. Wang, J. Song, M. Zheng, H. Sun, H. Wei, W. Xiao, R. Sun, Z. Tian, High NKG2A expression contributes to NK cell exhaustion and predicts a poor prognosis of patients with liver cancer. Oncoimmunol 6, e1264562 (2017) C. Sun, J. Xu, Q. Huang, M. Huang, H. Wen, C. Zhang, J. Wang, J. Song, M. Zheng, H. Sun, H. Wei, W. Xiao, R. Sun, Z. Tian, High NKG2A expression contributes to NK cell exhaustion and predicts a poor prognosis of patients with liver cancer. Oncoimmunol 6, e1264562 (2017)
94.
go back to reference S. Platonova, J. Cherfils-Vicini, D. Damotte, L. Crozet, V. Vieillard, P. Validire, P. André, M.C. Dieu-Nosjean, M. Alifano, J.F. Régnard, W.H. Fridman, C. Sautès-Fridman, I. Cremer, Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res 71, 5412–5422 (2011)PubMed S. Platonova, J. Cherfils-Vicini, D. Damotte, L. Crozet, V. Vieillard, P. Validire, P. André, M.C. Dieu-Nosjean, M. Alifano, J.F. Régnard, W.H. Fridman, C. Sautès-Fridman, I. Cremer, Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res 71, 5412–5422 (2011)PubMed
95.
go back to reference M. Gillard-Bocquet, C. Caer, N. Cagnard, L. Crozet, M. Perez, W.H. Fridman, C. Sautès-Fridman, I. Cremer, Lung tumor microenvironment induces specific gene expression signature in intratumoral NK cells. Front Immunol 4, 19 (2013)PubMedPubMedCentral M. Gillard-Bocquet, C. Caer, N. Cagnard, L. Crozet, M. Perez, W.H. Fridman, C. Sautès-Fridman, I. Cremer, Lung tumor microenvironment induces specific gene expression signature in intratumoral NK cells. Front Immunol 4, 19 (2013)PubMedPubMedCentral
96.
go back to reference E. Mamessier, A. Sylvain, M.L. Thibult, G. Houvenaeghel, J. Jacquemier, R. Castellano, A. Gonçalves, P. André, F. Romagné, G. Thibault, P. Viens, D. Birnbaum, F. Bertucci, A. Moretta, D. Olive, Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest 121, 3609–3622 (2011)PubMedPubMedCentral E. Mamessier, A. Sylvain, M.L. Thibult, G. Houvenaeghel, J. Jacquemier, R. Castellano, A. Gonçalves, P. André, F. Romagné, G. Thibault, P. Viens, D. Birnbaum, F. Bertucci, A. Moretta, D. Olive, Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest 121, 3609–3622 (2011)PubMedPubMedCentral
97.
go back to reference K. Stringaris, T. Sekine, A. Khoder, A. Alsuliman, B. Razzaghi, R. Sargeant, J. Pavlu, G. Brisley, H. de Lavallade, A. Sarvaria, D. Marin, S. Mielke, J.F. Apperley, E.J. Shpall, A.J. Barrett, K. Rezvani, Leukemia-induced phenotypic and functional defects in natural killer cells predict failure to achieve remission in acute myeloid leukemia. Haematologica 99, 836–847 (2014) K. Stringaris, T. Sekine, A. Khoder, A. Alsuliman, B. Razzaghi, R. Sargeant, J. Pavlu, G. Brisley, H. de Lavallade, A. Sarvaria, D. Marin, S. Mielke, J.F. Apperley, E.J. Shpall, A.J. Barrett, K. Rezvani, Leukemia-induced phenotypic and functional defects in natural killer cells predict failure to achieve remission in acute myeloid leukemia. Haematologica 99, 836–847 (2014)
98.
go back to reference P. André, C. Denis, C. Soulas, C. Bourbon-Caillet, J. Lopez, T. Arnoux, M. Bléry, C. Bonnafous, L. Gauthier, A. Morel, B. Rossi, R. Remark, V. Breso, E. Bonnet, G. Habif, S. Guia, A. I. Lalanne, C. Hoffmann, O. Lantz, J. Fayette, A. Boyer-Chammard, R. Zerbib, P. Dodion, H. Ghadially, M. Jure-Kunkel, Y. Morel, R. Herbst, E. Narni-Mancinelli, R. B. Cohen, E. Vivier, Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 175, 1731–1743 (2018) P. André, C. Denis, C. Soulas, C. Bourbon-Caillet, J. Lopez, T. Arnoux, M. Bléry, C. Bonnafous, L. Gauthier, A. Morel, B. Rossi, R. Remark, V. Breso, E. Bonnet, G. Habif, S. Guia, A. I. Lalanne, C. Hoffmann, O. Lantz, J. Fayette, A. Boyer-Chammard, R. Zerbib, P. Dodion, H. Ghadially, M. Jure-Kunkel, Y. Morel, R. Herbst, E. Narni-Mancinelli, R. B. Cohen, E. Vivier, Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 175, 1731–1743 (2018)
99.
go back to reference M. Benevolo, M. Mottolese, E. Tremante, F. Rollo, M.G. Diodoro, C. Ercolani, I. Sperduti, E. Lo Monaco, M. Cosimelli, P. Giacomini, High expression of HLA-E in colorectal carcinoma is associated with a favorable prognosis. J Transl Med 9, 184 (2011)PubMedPubMedCentral M. Benevolo, M. Mottolese, E. Tremante, F. Rollo, M.G. Diodoro, C. Ercolani, I. Sperduti, E. Lo Monaco, M. Cosimelli, P. Giacomini, High expression of HLA-E in colorectal carcinoma is associated with a favorable prognosis. J Transl Med 9, 184 (2011)PubMedPubMedCentral
100.
go back to reference T. van Hall, P. André, A. Horowitz, D. F. Ruan, L. Borst, R. Zerbib, E. Narni-Mancinelli, S. H van der Burg, E. Vivier. Monalizumab: Inhibiting the novel immune checkpoint NKG2A. J ImmunoTher Cancer 7, 263 (2019) T. van Hall, P. André, A. Horowitz, D. F. Ruan, L. Borst, R. Zerbib, E. Narni-Mancinelli, S. H van der Burg, E. Vivier. Monalizumab: Inhibiting the novel immune checkpoint NKG2A. J ImmunoTher Cancer 7, 263 (2019)
101.
go back to reference B.C. Creelan, S.J. Antonia, The NKG2A immune checkpoint — A new direction in cancer immunotherapy. Nat Rev Clin Oncol 16, 277 (2019)PubMed B.C. Creelan, S.J. Antonia, The NKG2A immune checkpoint — A new direction in cancer immunotherapy. Nat Rev Clin Oncol 16, 277 (2019)PubMed
102.
go back to reference C. Pfeifer, A.J. Highton, S. Peine, J. Sauter, A.H. Schmidt, M.J. Bunders, M. Altfeld, C. Körner, Natural killer cell education is associated with a distinct glycolytic profile. Front Immunol 9, 3020 (2018)PubMedPubMedCentral C. Pfeifer, A.J. Highton, S. Peine, J. Sauter, A.H. Schmidt, M.J. Bunders, M. Altfeld, C. Körner, Natural killer cell education is associated with a distinct glycolytic profile. Front Immunol 9, 3020 (2018)PubMedPubMedCentral
103.
go back to reference Y. Liu, Y. Cheng, Y. Xu, Z. Wang, X. Du, C. Li, J. Peng, L. Gao, X. Liang, C. Ma, Increased expression of programmed cell death protein 1 on NK cells inhibits NK-cell-mediated anti-tumor function and indicates poor prognosis in digestive cancers. Oncogene 36, 6143–6153 (2017) Y. Liu, Y. Cheng, Y. Xu, Z. Wang, X. Du, C. Li, J. Peng, L. Gao, X. Liang, C. Ma, Increased expression of programmed cell death protein 1 on NK cells inhibits NK-cell-mediated anti-tumor function and indicates poor prognosis in digestive cancers. Oncogene 36, 6143–6153 (2017)
104.
go back to reference J. Hsu, J.J. Hodgins, M. Marathe, C.J. Nicolai, M.C. Bourgeois-Daigneault, T.N. Trevino, C.S. Azimi, A.K. Scheer, H.E. Randolph, T.W. Thompson, L. Zhang, A. Iannello, N. Mathur, K.E. Jardine, G.A. Kirn, J.C. Bell, M.W. McBurney, D.H. Raulet, M. Ardolino, Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest 128, 4654–4668 (2018)PubMedPubMedCentral J. Hsu, J.J. Hodgins, M. Marathe, C.J. Nicolai, M.C. Bourgeois-Daigneault, T.N. Trevino, C.S. Azimi, A.K. Scheer, H.E. Randolph, T.W. Thompson, L. Zhang, A. Iannello, N. Mathur, K.E. Jardine, G.A. Kirn, J.C. Bell, M.W. McBurney, D.H. Raulet, M. Ardolino, Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest 128, 4654–4668 (2018)PubMedPubMedCentral
105.
go back to reference D. Sarhan, K.L. Hippen, A. Lemire, S. Hying, X. Luo, T. Lenvik, J. Curtsinger, Z. Davis, B. Zhang, S. Cooley, F. Cichocki, B.R. Blazar, J.S. Miller, Adaptive NK cells resist regulatory T-cell suppression driven by IL37. Cancer Immunol Res 6, 766–775 (2018) D. Sarhan, K.L. Hippen, A. Lemire, S. Hying, X. Luo, T. Lenvik, J. Curtsinger, Z. Davis, B. Zhang, S. Cooley, F. Cichocki, B.R. Blazar, J.S. Miller, Adaptive NK cells resist regulatory T-cell suppression driven by IL37. Cancer Immunol Res 6, 766–775 (2018)
106.
go back to reference Y. Huang, Z. Chen, J.H. Jang, M.S. Baig, G. Bertolet, C. Schroeder, S. Huang, Q. Hu, Y. Zhao, D.E. Lewis, L. Qin, M.X. Zhu, D. Liu, PD-1 blocks lytic granule polarization with concomitant impairment of integrin outside-in signaling in the natural killer cell immunological synapse. J Allergy Clin Immunol 142, 1311–1321 (2018)PubMedPubMedCentral Y. Huang, Z. Chen, J.H. Jang, M.S. Baig, G. Bertolet, C. Schroeder, S. Huang, Q. Hu, Y. Zhao, D.E. Lewis, L. Qin, M.X. Zhu, D. Liu, PD-1 blocks lytic granule polarization with concomitant impairment of integrin outside-in signaling in the natural killer cell immunological synapse. J Allergy Clin Immunol 142, 1311–1321 (2018)PubMedPubMedCentral
107.
go back to reference A. Beldi-Ferchiou, M. Lambert, S. Dogniaux, F. Vély, E. Vivier, D. Olive, S. Dupuy, F. Levasseur, D. Zucman, C. Lebbé, D. Sène, C. Hivroz, S. Caillat-Zucman, PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget 7, 72961–72977 (2016) A. Beldi-Ferchiou, M. Lambert, S. Dogniaux, F. Vély, E. Vivier, D. Olive, S. Dupuy, F. Levasseur, D. Zucman, C. Lebbé, D. Sène, C. Hivroz, S. Caillat-Zucman, PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget 7, 72961–72977 (2016)
108.
go back to reference S.J. Blake, W.C. Dougall, J.J. Miles, M.W.L. Teng, M.J. Smyth, Molecular pathways: Targeting CD96 and TIGIT for Cancer immunotherapy. Clin Cancer Res 22, 5183–5188 (2016)PubMed S.J. Blake, W.C. Dougall, J.J. Miles, M.W.L. Teng, M.J. Smyth, Molecular pathways: Targeting CD96 and TIGIT for Cancer immunotherapy. Clin Cancer Res 22, 5183–5188 (2016)PubMed
109.
go back to reference N. Stanietsky, H. Simic, J. Arapovic, A. Toporik, O. Levy, A. Novik, Z. Levine, M. Beiman, L. Dassa, H. Achdout, N. Stern-Ginossar, P. Tsukerman, S. Jonjic, O. Mandelboim, The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci U S A 106, 17858–17863 (2009)PubMedPubMedCentral N. Stanietsky, H. Simic, J. Arapovic, A. Toporik, O. Levy, A. Novik, Z. Levine, M. Beiman, L. Dassa, H. Achdout, N. Stern-Ginossar, P. Tsukerman, S. Jonjic, O. Mandelboim, The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci U S A 106, 17858–17863 (2009)PubMedPubMedCentral
110.
go back to reference W.C. Dougall, S. Kurtulus, M.J. Smyth, A.C. Anderson, TIGIT and CD96: New checkpoint receptor targets for cancer immunotherapy. Immunol Rev 276, 112–120 (2017)PubMed W.C. Dougall, S. Kurtulus, M.J. Smyth, A.C. Anderson, TIGIT and CD96: New checkpoint receptor targets for cancer immunotherapy. Immunol Rev 276, 112–120 (2017)PubMed
111.
go back to reference Q. Zhang, J. Bi, X. Zheng, Y. Chen, H. Wang, W. Wu, Z. Wang, Q. Wu, H. Peng, H. Wei, R. Sun, Z. Tian, Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol 19, 723–732 (2018)PubMed Q. Zhang, J. Bi, X. Zheng, Y. Chen, H. Wang, W. Wu, Z. Wang, Q. Wu, H. Peng, H. Wei, R. Sun, Z. Tian, Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol 19, 723–732 (2018)PubMed
112.
go back to reference K.F. Stengel, K. Harden-Bowles, X. Yu, L. Rouge, J. Yin, L. Comps-Agrar, C. Wiesmann, J.F. Bazan, D.L. Eaton, J.L. Grogan, Structure of TIGIT immunoreceptor bound to poliovirus receptor reveals a cell-cell adhesion and signaling mechanism that requires cis-trans receptor clustering. Proc Natl Acad Sci U S A 109, 5399–5404 (2012)PubMedPubMedCentral K.F. Stengel, K. Harden-Bowles, X. Yu, L. Rouge, J. Yin, L. Comps-Agrar, C. Wiesmann, J.F. Bazan, D.L. Eaton, J.L. Grogan, Structure of TIGIT immunoreceptor bound to poliovirus receptor reveals a cell-cell adhesion and signaling mechanism that requires cis-trans receptor clustering. Proc Natl Acad Sci U S A 109, 5399–5404 (2012)PubMedPubMedCentral
113.
go back to reference S. Liu, H. Zhang, M. Li, D. Hu, C. Li, B. Ge, B. Jin, Z. Fan, Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells. Cell Death Differ 20, 456–464 (2013)PubMed S. Liu, H. Zhang, M. Li, D. Hu, C. Li, B. Ge, B. Jin, Z. Fan, Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells. Cell Death Differ 20, 456–464 (2013)PubMed
114.
go back to reference A.M. Farkas, F. Audenet, H. Anastos, M. Galsky, J. Sfakianos, N. Bhardwaj, Tim-3 and TIGIT mark natural killer cells susceptible to effector dysfunction in human bladder cancer. J Immunol 200, 124.14 (2018) A.M. Farkas, F. Audenet, H. Anastos, M. Galsky, J. Sfakianos, N. Bhardwaj, Tim-3 and TIGIT mark natural killer cells susceptible to effector dysfunction in human bladder cancer. J Immunol 200, 124.14 (2018)
115.
go back to reference L.E. Lucca, P.P. Axisa, E.R. Singer, N.M. Nolan, M. Dominguez-Villar, D.A. Hafler, TIGIT signaling restores suppressor function of Th1 Tregs. JCI Insight 4, e124427 (2019)PubMedCentral L.E. Lucca, P.P. Axisa, E.R. Singer, N.M. Nolan, M. Dominguez-Villar, D.A. Hafler, TIGIT signaling restores suppressor function of Th1 Tregs. JCI Insight 4, e124427 (2019)PubMedCentral
116.
go back to reference M. Molgora, E. Bonavita, A. Ponzetta, F. Riva, M. Barbagallo, S. Jaillon, B. Popović, G. Bernardini, E. Magrini, F. Gianni, S. Zelenay, S. Jonjić, A. Santoni, C. Garlanda, A. Mantovani, IL-1R8 is a checkpoint in NK cells regulating anti-tumor and anti-viral activity. Nature 551, 110–114 (2017) M. Molgora, E. Bonavita, A. Ponzetta, F. Riva, M. Barbagallo, S. Jaillon, B. Popović, G. Bernardini, E. Magrini, F. Gianni, S. Zelenay, S. Jonjić, A. Santoni, C. Garlanda, A. Mantovani, IL-1R8 is a checkpoint in NK cells regulating anti-tumor and anti-viral activity. Nature 551, 110–114 (2017)
117.
go back to reference L. Long, X. Zhang, F. Chen, Q. Pan, P. Phiphatwatchara, Y. Zeng, H. Chen, The promising immune checkpoint LAG-3: From tumor microenvironment to cancer immunotherapy. Genes Cancer 9, 176–189 (2018)PubMedPubMedCentral L. Long, X. Zhang, F. Chen, Q. Pan, P. Phiphatwatchara, Y. Zeng, H. Chen, The promising immune checkpoint LAG-3: From tumor microenvironment to cancer immunotherapy. Genes Cancer 9, 176–189 (2018)PubMedPubMedCentral
118.
go back to reference B. Huard, M. Tournier, F. Triebel, LAG-3 does not define a specific mode of natural killing in human. Immunol Lett 61, 109–112 (1998)PubMed B. Huard, M. Tournier, F. Triebel, LAG-3 does not define a specific mode of natural killing in human. Immunol Lett 61, 109–112 (1998)PubMed
119.
go back to reference A.C. Anderson, N. Joller, V.K. Kuchroo, Lag-3, Tim-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity 44, 989–1004 (2016) A.C. Anderson, N. Joller, V.K. Kuchroo, Lag-3, Tim-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity 44, 989–1004 (2016)
120.
go back to reference P.A. Ascierto, I. Melero, S. Bhatia, P. Bono, R.E. Sanborn, E.J. Lipson, M.K. Callahan, T. Gajewski, C.A. Gomez-Roca, F.S. Hodi, G. Curigliano, M. Nyakas, M. Preusser, Y. Koguchi, M. Maurer, R. Clynes, P. Mitra, S. Suryawanshi, E. Muñoz-Couselo, Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy. J Clin Oncol 35, 9520–9520 (2017) P.A. Ascierto, I. Melero, S. Bhatia, P. Bono, R.E. Sanborn, E.J. Lipson, M.K. Callahan, T. Gajewski, C.A. Gomez-Roca, F.S. Hodi, G. Curigliano, M. Nyakas, M. Preusser, Y. Koguchi, M. Maurer, R. Clynes, P. Mitra, S. Suryawanshi, E. Muñoz-Couselo, Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy. J Clin Oncol 35, 9520–9520 (2017)
121.
go back to reference J.M. Wang, Y.Q. Cheng, L. Shi, R.S. Ying, X.Y. Wu, G.Y. Li, J.P. Moorman, Z.Q. Yao, KLRG1 negatively regulates natural killer cell functions through the Akt pathway in individuals with chronic hepatitis C virus infection. J Virol 87, 11626–11636 (2013)PubMedPubMedCentral J.M. Wang, Y.Q. Cheng, L. Shi, R.S. Ying, X.Y. Wu, G.Y. Li, J.P. Moorman, Z.Q. Yao, KLRG1 negatively regulates natural killer cell functions through the Akt pathway in individuals with chronic hepatitis C virus infection. J Virol 87, 11626–11636 (2013)PubMedPubMedCentral
122.
go back to reference B. Müller-Durovic, A. Lanna, L.P. Covre, R.S. Mills, S.M. Henson, A.N. Akbar, Killer cell lectin-like receptor G1 (KLRG1) inhibits NK cell function through activation of AMP-activated protein kinase. J Immunol 197, 2891–2899 (2016)PubMedPubMedCentral B. Müller-Durovic, A. Lanna, L.P. Covre, R.S. Mills, S.M. Henson, A.N. Akbar, Killer cell lectin-like receptor G1 (KLRG1) inhibits NK cell function through activation of AMP-activated protein kinase. J Immunol 197, 2891–2899 (2016)PubMedPubMedCentral
123.
go back to reference R. B. Delconte, T. B. Kolesnik, L. F. Dagley, J. Rautela, W. Shi, E. M. Putz, K. Stannard, J. G. Zhang, C. Teh, M. Firth, T. Ushiki, C. E. Andoniou, M. A. Degli-Esposti, P. P. Sharp, C. E. Sanvitale, G. Infusini, N. P. D. Liau, E. M. Linossi, C. J. Burns, S. Carotta, D. H. D. Gray, C. Seillet, D. S. Hutchinson, G. T. Belz, A. I. Webb, W. S. Alexander, S. S. Li, A. N. Bullock, J. J. Babon, M. J. Smyth, S. E. Nicholson, N. D. Huntington, CIS is a potent checkpoint in NK cell-mediated tumor immunity. Nat Immunol 17, 816–824 (2016) R. B. Delconte, T. B. Kolesnik, L. F. Dagley, J. Rautela, W. Shi, E. M. Putz, K. Stannard, J. G. Zhang, C. Teh, M. Firth, T. Ushiki, C. E. Andoniou, M. A. Degli-Esposti, P. P. Sharp, C. E. Sanvitale, G. Infusini, N. P. D. Liau, E. M. Linossi, C. J. Burns, S. Carotta, D. H. D. Gray, C. Seillet, D. S. Hutchinson, G. T. Belz, A. I. Webb, W. S. Alexander, S. S. Li, A. N. Bullock, J. J. Babon, M. J. Smyth, S. E. Nicholson, N. D. Huntington, CIS is a potent checkpoint in NK cell-mediated tumor immunity. Nat Immunol 17, 816–824 (2016)
124.
go back to reference E.M. Putz, C. Guillerey, K. Kos, K. Stannard, K. Miles, R.B. Delconte, K. Takeda, S.E. Nicholson, N.D. Huntington, M.J. Smyth, Targeting cytokine signaling checkpoint CIS activates NK cells to protect from tumor initiation and metastasis. OncoImmunol 6, e1267892 (2017) E.M. Putz, C. Guillerey, K. Kos, K. Stannard, K. Miles, R.B. Delconte, K. Takeda, S.E. Nicholson, N.D. Huntington, M.J. Smyth, Targeting cytokine signaling checkpoint CIS activates NK cells to protect from tumor initiation and metastasis. OncoImmunol 6, e1267892 (2017)
125.
go back to reference E.C. So, A. Khaladj-Ghom, Y. Ji, J. Amin, Y. Song, E. Burch, H. Zhou, H. Sun, S. Chen, S. Bentzen, R. Hertzano, X. Zhang, S.E. Strome, NK cell expression of Tim-3: First impressions matter. Immunobiology. 224, 362–370 (2019)PubMed E.C. So, A. Khaladj-Ghom, Y. Ji, J. Amin, Y. Song, E. Burch, H. Zhou, H. Sun, S. Chen, S. Bentzen, R. Hertzano, X. Zhang, S.E. Strome, NK cell expression of Tim-3: First impressions matter. Immunobiology. 224, 362–370 (2019)PubMed
126.
go back to reference L.C. Ndhlovu, S. Lopez-Vergès, J.D. Barbour, R.B. Jones, A.R. Jha, B.R. Long, E.C. Schoeffler, T. Fujita, D.F. Nixon, L.L. Lanier, Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood 119, 3734–3743 (2012) L.C. Ndhlovu, S. Lopez-Vergès, J.D. Barbour, R.B. Jones, A.R. Jha, B.R. Long, E.C. Schoeffler, T. Fujita, D.F. Nixon, L.L. Lanier, Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood 119, 3734–3743 (2012)
127.
go back to reference L.Y. Xu, D.D. Chen, J.Y. He, C.C. Lu, X.G. Liu, H.B. Le, C.Y. Wang, Y.K. Zhang, Tim-3 expression by peripheral natural killer cells and natural killer T cells increases in patients with lung cancer--reduction after surgical resection. Asian Pac J Cancer Prev 15, 9945–9948 (2014)PubMed L.Y. Xu, D.D. Chen, J.Y. He, C.C. Lu, X.G. Liu, H.B. Le, C.Y. Wang, Y.K. Zhang, Tim-3 expression by peripheral natural killer cells and natural killer T cells increases in patients with lung cancer--reduction after surgical resection. Asian Pac J Cancer Prev 15, 9945–9948 (2014)PubMed
128.
go back to reference L. Xu, Y. Huang, L. Tan, W. Yu, D. Chen, C. Lu, J. He, G. Wu, X. Liu, Y. Zhang, Increased Tim-3 expression in peripheral NK cells predicts a poorer prognosis and Tim-3 blockade improves NK cell-mediated cytotoxicity in human lung adenocarcinoma. Int Immunopharmacol 29, 635–641 (2015)PubMed L. Xu, Y. Huang, L. Tan, W. Yu, D. Chen, C. Lu, J. He, G. Wu, X. Liu, Y. Zhang, Increased Tim-3 expression in peripheral NK cells predicts a poorer prognosis and Tim-3 blockade improves NK cell-mediated cytotoxicity in human lung adenocarcinoma. Int Immunopharmacol 29, 635–641 (2015)PubMed
129.
go back to reference E. Batlle, J. Massagué, Transforming growth factor-β signaling in immunity and Cancer. Immunity 50, 924–940 (2019) E. Batlle, J. Massagué, Transforming growth factor-β signaling in immunity and Cancer. Immunity 50, 924–940 (2019)
130.
go back to reference S. Colak, P. Ten Dijke, Targeting TGF-β signaling in Cancer. Trends Cancer 3, 56–71 (2017)PubMed S. Colak, P. Ten Dijke, Targeting TGF-β signaling in Cancer. Trends Cancer 3, 56–71 (2017)PubMed
131.
go back to reference R. Trotta, J. Dal Col, J. Yu, D. Ciarlariello, B. Thomas, X. Zhang, J. Allard, M. Wei, H. Mao, J.C. Byrd, D. Perrotti, M.A. Caligiuri, TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol 181, 3784–3792 (2008)PubMedPubMedCentral R. Trotta, J. Dal Col, J. Yu, D. Ciarlariello, B. Thomas, X. Zhang, J. Allard, M. Wei, H. Mao, J.C. Byrd, D. Perrotti, M.A. Caligiuri, TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol 181, 3784–3792 (2008)PubMedPubMedCentral
132.
go back to reference B. Han, F.Y. Mao, Y.L. Zhao, Y.P. Lv, Y.S. Teng, M. Duan, W. Chen, P. Cheng, T.T. Wang, Z.Y. Liang, J.Y. Zhang, Y.G. Liu, G. Guo, Q.M. Zou, Y. Zhuang, L.S. Peng, Altered NKp30, NKp46, NKG2D, and DNAM-1 expression on circulating NK cells is associated with tumor progression in human gastric cancer. J Immunol Res 2018, 6248590 (2018) B. Han, F.Y. Mao, Y.L. Zhao, Y.P. Lv, Y.S. Teng, M. Duan, W. Chen, P. Cheng, T.T. Wang, Z.Y. Liang, J.Y. Zhang, Y.G. Liu, G. Guo, Q.M. Zou, Y. Zhuang, L.S. Peng, Altered NKp30, NKp46, NKG2D, and DNAM-1 expression on circulating NK cells is associated with tumor progression in human gastric cancer. J Immunol Res 2018, 6248590 (2018)
133.
go back to reference Y. Gao, F. Souza-Fonseca-Guimaraes, T. Bald, S.S. Ng, A. Young, S.F. Ngiow, J. Rautela, J. Straube, N. Waddell, S.J. Blake, J. Yan, L. Bartholin, J.S. Lee, E. Vivier, K. Takeda, M. Messaoudene, L. Zitvogel, M.W.L. Teng, G.T. Belz, C.R. Engwerda, N.D. Huntington, K. Nakamura, M. Hölzel, M.J. Smyth, Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat Immunol 18, 1004–1015 (2017)PubMed Y. Gao, F. Souza-Fonseca-Guimaraes, T. Bald, S.S. Ng, A. Young, S.F. Ngiow, J. Rautela, J. Straube, N. Waddell, S.J. Blake, J. Yan, L. Bartholin, J.S. Lee, E. Vivier, K. Takeda, M. Messaoudene, L. Zitvogel, M.W.L. Teng, G.T. Belz, C.R. Engwerda, N.D. Huntington, K. Nakamura, M. Hölzel, M.J. Smyth, Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat Immunol 18, 1004–1015 (2017)PubMed
134.
go back to reference A. Dahmani, J.S. Delisle, TGF-β in T cell biology: Implications for cancer immunotherapy. Cancers 10, 6 (2018) A. Dahmani, J.S. Delisle, TGF-β in T cell biology: Implications for cancer immunotherapy. Cancers 10, 6 (2018)
135.
go back to reference H. Li, Y. Han, Q. Guo, M. Zhang, X. Cao, Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol 182, 240–249 (2009)PubMed H. Li, Y. Han, Q. Guo, M. Zhang, X. Cao, Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol 182, 240–249 (2009)PubMed
136.
go back to reference Z. Li, Y. Pang, S.K. Gara, B.R. Achyut, C. Heger, P.K. Goldsmith, S. Lonning, L. Yang, Gr-1+CD11b+ cells are responsible for tumor promoting effect of TGF-β in breast cancer progression. Int J Cancer 131, 2584–2595 (2012)PubMedPubMedCentral Z. Li, Y. Pang, S.K. Gara, B.R. Achyut, C. Heger, P.K. Goldsmith, S. Lonning, L. Yang, Gr-1+CD11b+ cells are responsible for tumor promoting effect of TGF-β in breast cancer progression. Int J Cancer 131, 2584–2595 (2012)PubMedPubMedCentral
137.
go back to reference Z.G. Fridlender, J. Sun, S. Kim, V. Kapoor, G. Cheng, L. Ling, G.S. Worthen, S.M. Albelda, Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 16, 183–119 (2009)PubMedPubMedCentral Z.G. Fridlender, J. Sun, S. Kim, V. Kapoor, G. Cheng, L. Ling, G.S. Worthen, S.M. Albelda, Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 16, 183–119 (2009)PubMedPubMedCentral
138.
go back to reference J.C. Mbongue, D.A. Nicholas, T.W. Torrez, N.S. Kim, A.F. Firek, W.H.R. Langridge, The role of Indoleamine 2, 3-dioxygenase in immune suppression and autoimmunity. Vaccines 3, 703–729 (2015)PubMedPubMedCentral J.C. Mbongue, D.A. Nicholas, T.W. Torrez, N.S. Kim, A.F. Firek, W.H.R. Langridge, The role of Indoleamine 2, 3-dioxygenase in immune suppression and autoimmunity. Vaccines 3, 703–729 (2015)PubMedPubMedCentral
139.
go back to reference M. Foroutan, J. Cursons, S. Hediyeh-Zadeh, E.W. Thompson, M.J. Davis, A transcriptional program for detecting TGFβ-induced EMT in cancer. Mol Cancer Res 15, 619–631 (2017) M. Foroutan, J. Cursons, S. Hediyeh-Zadeh, E.W. Thompson, M.J. Davis, A transcriptional program for detecting TGFβ-induced EMT in cancer. Mol Cancer Res 15, 619–631 (2017)
140.
go back to reference S. Viel, A. Marçais, F.S.F. Guimaraes, R. Loftus, J. Rabilloud, M. Grau, S. Degouve, S. Djebali, A. Sanlaville, E. Charrier, J. Bienvenu, J.C. Marie, C. Caux, J. Marvel, L. Town, N.D. Huntington, L. Bartholin, D. Finlay, M.J. Smyth, T. Walzer, TGF-β inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci Signal 9, 415 (2016) S. Viel, A. Marçais, F.S.F. Guimaraes, R. Loftus, J. Rabilloud, M. Grau, S. Degouve, S. Djebali, A. Sanlaville, E. Charrier, J. Bienvenu, J.C. Marie, C. Caux, J. Marvel, L. Town, N.D. Huntington, L. Bartholin, D. Finlay, M.J. Smyth, T. Walzer, TGF-β inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci Signal 9, 415 (2016)
141.
go back to reference V. Zaiatz-Bittencourt, D.K. Finlay, C.M. Gardiner, Canonical TGF-β signaling pathway represses human NK cell metabolism. J Immunol 200, 3934–3941 (2018)PubMed V. Zaiatz-Bittencourt, D.K. Finlay, C.M. Gardiner, Canonical TGF-β signaling pathway represses human NK cell metabolism. J Immunol 200, 3934–3941 (2018)PubMed
142.
go back to reference L. Antonioli, C. Blandizzi, P. Pacher, G. Haskó, Immunity, inflammation and cancer: A leading role for adenosine. Nat Rev Cancer 13, 842–857 (2013)PubMed L. Antonioli, C. Blandizzi, P. Pacher, G. Haskó, Immunity, inflammation and cancer: A leading role for adenosine. Nat Rev Cancer 13, 842–857 (2013)PubMed
143.
go back to reference J. Stagg, M.J. Smyth, Extracellular adenosine triphosphate and adenosine in cancer. Oncogene 29, 5346–5358 (2010) J. Stagg, M.J. Smyth, Extracellular adenosine triphosphate and adenosine in cancer. Oncogene 29, 5346–5358 (2010)
144.
145.
go back to reference A. Young, D. Mittal, J. Stagg, M.J. Smyth, Targeting cancer-derived adenosine: New therapeutic approaches. Cancer Discov 4, 879–888 (2014)PubMed A. Young, D. Mittal, J. Stagg, M.J. Smyth, Targeting cancer-derived adenosine: New therapeutic approaches. Cancer Discov 4, 879–888 (2014)PubMed
146.
go back to reference A. Young, S.F. Ngiow, D.S. Barkauskas, E. Sult, C. Hay, S.J. Blake, Q. Huang, J. Liu, K. Takeda, M.W. Teng, Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses. Cancer Cell 30, 391–403 (2016)PubMed A. Young, S.F. Ngiow, D.S. Barkauskas, E. Sult, C. Hay, S.J. Blake, Q. Huang, J. Liu, K. Takeda, M.W. Teng, Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses. Cancer Cell 30, 391–403 (2016)PubMed
147.
go back to reference J. Wang, S. Matosevic, Adenosinergic signaling as a target for natural killer cell immunotherapy. J Mol Med 96, 903–913 (2018)PubMed J. Wang, S. Matosevic, Adenosinergic signaling as a target for natural killer cell immunotherapy. J Mol Med 96, 903–913 (2018)PubMed
148.
go back to reference A. Greenhough, H.J.M. Smartt, A.E. Moore, H.R. Roberts, A.C. Williams, C. Paraskeva, A. Kaidi, The COX-2/PGE2 pathway: Key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis 30, 377–386 (2009) A. Greenhough, H.J.M. Smartt, A.E. Moore, H.R. Roberts, A.C. Williams, C. Paraskeva, A. Kaidi, The COX-2/PGE2 pathway: Key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis 30, 377–386 (2009)
149.
150.
go back to reference S. Zelenay, A. G. van der Veen, J. P. Böttcher, K. J. Snelgrove, N. Rogers, S. E. Acton, P. Chakravarty, M. R. Girotti, R. Marais, S. A. Quezada, E. Sahai, C. Reis e Sousa, Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell 162, 1257–1270 (2015) S. Zelenay, A. G. van der Veen, J. P. Böttcher, K. J. Snelgrove, N. Rogers, S. E. Acton, P. Chakravarty, M. R. Girotti, R. Marais, S. A. Quezada, E. Sahai, C. Reis e Sousa, Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell 162, 1257–1270 (2015)
151.
go back to reference D. Wang, R.N. DuBois, The role of prostaglandin E(2) in tumor-associated immunosuppression. Trends Mol Med 22, 1–3 (2016)PubMed D. Wang, R.N. DuBois, The role of prostaglandin E(2) in tumor-associated immunosuppression. Trends Mol Med 22, 1–3 (2016)PubMed
152.
go back to reference J. Ke, Y. Yang, Q. Che, F. Jiang, H. Wang, Z. Chen, M. Zhu, H. Tong, H. Zhang, X. Yan, X. Wang, F. Wang, Y. Liu, C. Dai, X. Wan, Prostaglandin E2 (PGE2) promotes proliferation and invasion by enhancing SUMO-1 activity via EP4 receptor in endometrial cancer. Tumour Biol 37, 12203–12211 (2016)PubMedPubMedCentral J. Ke, Y. Yang, Q. Che, F. Jiang, H. Wang, Z. Chen, M. Zhu, H. Tong, H. Zhang, X. Yan, X. Wang, F. Wang, Y. Liu, C. Dai, X. Wan, Prostaglandin E2 (PGE2) promotes proliferation and invasion by enhancing SUMO-1 activity via EP4 receptor in endometrial cancer. Tumour Biol 37, 12203–12211 (2016)PubMedPubMedCentral
153.
go back to reference B.C. Lee, H.S. Kim, T.H. Shin, I. Kang, J.Y. Lee, J.J. Kim, H.K. Kang, Y. Seo, S. Lee, K.R. Yu, S.W. Choi, K.S. Kang, PGE2 maintains self-renewal of human adult stem cells via EP2-mediated autocrine signaling and its production is regulated by cell-to-cell contact. Sci Rep 6, 26298 (2016)PubMedPubMedCentral B.C. Lee, H.S. Kim, T.H. Shin, I. Kang, J.Y. Lee, J.J. Kim, H.K. Kang, Y. Seo, S. Lee, K.R. Yu, S.W. Choi, K.S. Kang, PGE2 maintains self-renewal of human adult stem cells via EP2-mediated autocrine signaling and its production is regulated by cell-to-cell contact. Sci Rep 6, 26298 (2016)PubMedPubMedCentral
155.
go back to reference C.H.M.J. Van Elssen, J. Vanderlocht, T. Oth, B.L.M.G. Senden-Gijsbers, W.T.V. Germeraad, G.M.J. Bos, Inflammation-restraining effects of prostaglandin E2 on natural killer-dendritic cell (NK-DC) interaction are imprinted during DC maturation. Blood 118, 2473–2482 (2011) C.H.M.J. Van Elssen, J. Vanderlocht, T. Oth, B.L.M.G. Senden-Gijsbers, W.T.V. Germeraad, G.M.J. Bos, Inflammation-restraining effects of prostaglandin E2 on natural killer-dendritic cell (NK-DC) interaction are imprinted during DC maturation. Blood 118, 2473–2482 (2011)
156.
go back to reference J. P. Böttcher, E. Bonavita, P. Chakravarty, H. Blees, M. Cabeza-Cabrerizo, S. Sammicheli, N. C. Rogers, E. Sahai, S. Zelenay, C. Reis e Sousa. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037 (2018) J. P. Böttcher, E. Bonavita, P. Chakravarty, H. Blees, M. Cabeza-Cabrerizo, S. Sammicheli, N. C. Rogers, E. Sahai, S. Zelenay, C. Reis e Sousa. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037 (2018)
157.
go back to reference M.Y. Jung, H.S. Kuehn, M. Beaven, D. Metcalfe, A. Gilfillan, Prostaglandin E2 induces activation of mTORC1 and mTORC2 in mast cells: Selective utilization of mTORC2 for the regulation of chemotaxis and mediator release. J Immunol 186, 151.6–151.6 (2011) M.Y. Jung, H.S. Kuehn, M. Beaven, D. Metcalfe, A. Gilfillan, Prostaglandin E2 induces activation of mTORC1 and mTORC2 in mast cells: Selective utilization of mTORC2 for the regulation of chemotaxis and mediator release. J Immunol 186, 151.6–151.6 (2011)
158.
go back to reference D. Holt, X. Ma, N. Kundu, P.D. Collin, A.M. Fulton, Modulation of host natural killer cell functions in breast cancer via prostaglandin E2 receptors EP2 and EP4. J Immunother 35, 179–188 (2012)PubMedPubMedCentral D. Holt, X. Ma, N. Kundu, P.D. Collin, A.M. Fulton, Modulation of host natural killer cell functions in breast cancer via prostaglandin E2 receptors EP2 and EP4. J Immunother 35, 179–188 (2012)PubMedPubMedCentral
159.
go back to reference S. Ganapathy-Kanniappan, J.F.H. Geschwind, Tumor glycolysis as a target for cancer therapy: Progress and prospect. Mol Cancer 12, 152 (2013)PubMedPubMedCentral S. Ganapathy-Kanniappan, J.F.H. Geschwind, Tumor glycolysis as a target for cancer therapy: Progress and prospect. Mol Cancer 12, 152 (2013)PubMedPubMedCentral
160.
go back to reference Z. Husain, Y. Huang, P. Seth, V.P. Sukhatme, Tumor-derived lactate modifies antitumor immune response: Effect on myeloid-derived suppressor cells and NK cells. J Immunol 191, 1486–1495 (2013)PubMed Z. Husain, Y. Huang, P. Seth, V.P. Sukhatme, Tumor-derived lactate modifies antitumor immune response: Effect on myeloid-derived suppressor cells and NK cells. J Immunol 191, 1486–1495 (2013)PubMed
161.
go back to reference A. Brand, K. Singer, G. E. Koehl, M. Kolitzus, G. Schoenhammer, A. Thiel, C. Matos, C. Bruss, S. Klobuch, K. Peter, M. Kastenberger, C. Bogdan, U. Schleicher, A. Mackensen, E. Ullrich, S. Fichtner-Feigl, R. Kesselring, M. Mack, U. Ritter, M. Schmid, C. Blank, K. Dettmer, P. J. Oefner, P. Hoffmann, S. Walenta, E. K. Geissler, J. Pouyssegur, A. Villunger, A. Steven, B. Seliger, S. Schreml, S. Haferkamp, E. Kohl, S. Karrer, M. Berneburg, W. Herr, W. Mueller-Klieser, K. Renner, M. Kreutz, LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab 24, 657–671 (2016) A. Brand, K. Singer, G. E. Koehl, M. Kolitzus, G. Schoenhammer, A. Thiel, C. Matos, C. Bruss, S. Klobuch, K. Peter, M. Kastenberger, C. Bogdan, U. Schleicher, A. Mackensen, E. Ullrich, S. Fichtner-Feigl, R. Kesselring, M. Mack, U. Ritter, M. Schmid, C. Blank, K. Dettmer, P. J. Oefner, P. Hoffmann, S. Walenta, E. K. Geissler, J. Pouyssegur, A. Villunger, A. Steven, B. Seliger, S. Schreml, S. Haferkamp, E. Kohl, S. Karrer, M. Berneburg, W. Herr, W. Mueller-Klieser, K. Renner, M. Kreutz, LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab 24, 657–671 (2016)
162.
go back to reference V. Huber, C. Camisaschi, A. Berzi, S. Ferro, L. Lugini, T. Triulzi, A. Tuccitto, E. Tagliabue, C. Castelli, L. Rivoltini, Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol 43, 74–89 (2017)PubMed V. Huber, C. Camisaschi, A. Berzi, S. Ferro, L. Lugini, T. Triulzi, A. Tuccitto, E. Tagliabue, C. Castelli, L. Rivoltini, Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol 43, 74–89 (2017)PubMed
163.
go back to reference N. Rohani, L. Hao, M.S. Alexis, B.A. Joughin, K. Krismer, M.N. Moufarrej, A.R. Soltis, D.A. Lauffenburger, M.B. Yaffe, C.B. Burge, S.N. Bhatia, F.B. Gertler, Acidification of tumor at stromal boundaries drives transcriptome alterations associated with aggressive phenotypes. Cancer Res 79, 1952–1966 (2019)PubMedPubMedCentral N. Rohani, L. Hao, M.S. Alexis, B.A. Joughin, K. Krismer, M.N. Moufarrej, A.R. Soltis, D.A. Lauffenburger, M.B. Yaffe, C.B. Burge, S.N. Bhatia, F.B. Gertler, Acidification of tumor at stromal boundaries drives transcriptome alterations associated with aggressive phenotypes. Cancer Res 79, 1952–1966 (2019)PubMedPubMedCentral
164.
go back to reference Y. Long, Z. Gao, X. Hu, F. Xiang, Z. Wu, J. Zhang, X. Han, L. Yin, J. Qin, L. Lan, F. Yin, Y. Wang, Downregulation of MCT4 for lactate exchange promotes the cytotoxicity of NK cells in breast carcinoma. Cancer Med 7, 4690–4700 (2018) Y. Long, Z. Gao, X. Hu, F. Xiang, Z. Wu, J. Zhang, X. Han, L. Yin, J. Qin, L. Lan, F. Yin, Y. Wang, Downregulation of MCT4 for lactate exchange promotes the cytotoxicity of NK cells in breast carcinoma. Cancer Med 7, 4690–4700 (2018)
165.
go back to reference E. Wennerberg, V Kremer, R. Childs, A. Lundqvist. CXCL10-induced migration of adoptively transferred human natural killer cells toward solid tumors causes regression of tumor growth in vivo. Cancer Immunol Immunother 64, 225–235 (2015) E. Wennerberg, V Kremer, R. Childs, A. Lundqvist. CXCL10-induced migration of adoptively transferred human natural killer cells toward solid tumors causes regression of tumor growth in vivo. Cancer Immunol Immunother 64, 225–235 (2015)
166.
go back to reference V. Kremer, M.A. Ligtenberg, R. Zendehdel, C. Seitz, A. Duivenvoorden, E. Wennerberg, E. Colón, A.H. Scherman-Plogell, A. Lundqvist, Genetic engineering of human NK cells to express CXCR2 improves migration to renal cell carcinoma. J Immunother Cancer 5, 73 (2017)PubMedPubMedCentral V. Kremer, M.A. Ligtenberg, R. Zendehdel, C. Seitz, A. Duivenvoorden, E. Wennerberg, E. Colón, A.H. Scherman-Plogell, A. Lundqvist, Genetic engineering of human NK cells to express CXCR2 improves migration to renal cell carcinoma. J Immunother Cancer 5, 73 (2017)PubMedPubMedCentral
167.
go back to reference M. Wendel, I.E. Galani, E. Suri-Payer, A. Cerwenka, Natural killer cell accumulation in tumors is dependent on IFN-gamma and CXCR3 ligands. Cancer Res 68, 8437–8445 (2008)PubMed M. Wendel, I.E. Galani, E. Suri-Payer, A. Cerwenka, Natural killer cell accumulation in tumors is dependent on IFN-gamma and CXCR3 ligands. Cancer Res 68, 8437–8445 (2008)PubMed
168.
go back to reference A. Brech, T. Ahlquist, R.A. Lothe, H. Stenmark, Autophagy in tumour suppression and promotion. Mol Oncol 3, 366–375 (2009)PubMedPubMedCentral A. Brech, T. Ahlquist, R.A. Lothe, H. Stenmark, Autophagy in tumour suppression and promotion. Mol Oncol 3, 366–375 (2009)PubMedPubMedCentral
169.
go back to reference A.C. Kimmelman, E. White, Autophagy and Tumor Metabolism Cell Metab 25, 1037–1043 (2017)PubMed A.C. Kimmelman, E. White, Autophagy and Tumor Metabolism Cell Metab 25, 1037–1043 (2017)PubMed
170.
go back to reference E.Y. Liu, K.M. Ryan, Autophagy and cancer--issues we need to digest. J Cell Sci 125, 2349–2358 (2012)PubMed E.Y. Liu, K.M. Ryan, Autophagy and cancer--issues we need to digest. J Cell Sci 125, 2349–2358 (2012)PubMed
171.
go back to reference T. Mgrditchian, T. Arakelian, J. Paggetti, M.Z. Noman, E. Viry, E. Moussay, K. Van Moer, S. Kreis, C. Guerin, S. Buart, Targeting autophagy inhibits melanoma growth by enhancing NK cells infiltration in a CCL5-dependent manner. Proc Nat Acad Sci 114, E9271–E9279 (2017)PubMed T. Mgrditchian, T. Arakelian, J. Paggetti, M.Z. Noman, E. Viry, E. Moussay, K. Van Moer, S. Kreis, C. Guerin, S. Buart, Targeting autophagy inhibits melanoma growth by enhancing NK cells infiltration in a CCL5-dependent manner. Proc Nat Acad Sci 114, E9271–E9279 (2017)PubMed
172.
go back to reference I.K. Choi, R. Strauss, M. Richter, C.O. Yun, A. Lieber, Strategies to increase drug penetration in solid tumors. Front Oncol 3, 193 (2013)PubMedPubMedCentral I.K. Choi, R. Strauss, M. Richter, C.O. Yun, A. Lieber, Strategies to increase drug penetration in solid tumors. Front Oncol 3, 193 (2013)PubMedPubMedCentral
173.
go back to reference Q.T. Le, G. Shi, H. Cao, D.W. Nelson, Y. Wang, E.Y. Chen, S. Zhao, C. Kong, D. Richardson, K.J. O’Byrne, A.J. Giaccia, A.C. Koong, Galectin-1: A link between tumor hypoxia and tumor immune privilege. J Clin Oncol 23, 8932–8941 (2005)PubMed Q.T. Le, G. Shi, H. Cao, D.W. Nelson, Y. Wang, E.Y. Chen, S. Zhao, C. Kong, D. Richardson, K.J. O’Byrne, A.J. Giaccia, A.C. Koong, Galectin-1: A link between tumor hypoxia and tumor immune privilege. J Clin Oncol 23, 8932–8941 (2005)PubMed
174.
go back to reference V. Sundblad, L.G. Morosi, J.R. Geffner, G.A. Rabinovich, Galectin-1: A Jack-of-all-trades in the resolution of acute and chronic inflammation. J Immunol 199, 3721–3730 (2017)PubMed V. Sundblad, L.G. Morosi, J.R. Geffner, G.A. Rabinovich, Galectin-1: A Jack-of-all-trades in the resolution of acute and chronic inflammation. J Immunol 199, 3721–3730 (2017)PubMed
175.
go back to reference L. Astorgues-Xerri, M.E. Riveiro, A. Tijeras-Raballand, M. Serova, C. Neuzillet, S. Albert, E. Raymond, S. Faivre, Unraveling galectin-1 as a novel therapeutic target for cancer. Cancer Treat Rev 40, 307–319 (2014)PubMed L. Astorgues-Xerri, M.E. Riveiro, A. Tijeras-Raballand, M. Serova, C. Neuzillet, S. Albert, E. Raymond, S. Faivre, Unraveling galectin-1 as a novel therapeutic target for cancer. Cancer Treat Rev 40, 307–319 (2014)PubMed
176.
go back to reference R. Wu, T. Wu, K. Wang, S. Luo, Z. Chen, M. Fan, D. Xue, H. Lu, Q. Zhuang, X. Xu, Prognostic significance of galectin-1 expression in patients with cancer: A meta-analysis. Cancer Cell Int 18, 108 (2018)PubMedPubMedCentral R. Wu, T. Wu, K. Wang, S. Luo, Z. Chen, M. Fan, D. Xue, H. Lu, Q. Zhuang, X. Xu, Prognostic significance of galectin-1 expression in patients with cancer: A meta-analysis. Cancer Cell Int 18, 108 (2018)PubMedPubMedCentral
177.
go back to reference J.M. Cousin, M.J. Cloninger, The role of Galectin-1 in Cancer progression, and synthetic multivalent Systems for the Study of Galectin-1. Int J Mol Sci 17, 1566 (2016)PubMedCentral J.M. Cousin, M.J. Cloninger, The role of Galectin-1 in Cancer progression, and synthetic multivalent Systems for the Study of Galectin-1. Int J Mol Sci 17, 1566 (2016)PubMedCentral
178.
go back to reference F.C. Chou, H.Y. Chen, C.C. Kuo, H.K. Sytwu, Role of galectins in tumors and in clinical immunotherapy. Int J Mol Sci 19, 430 (2018)PubMedCentral F.C. Chou, H.Y. Chen, C.C. Kuo, H.K. Sytwu, Role of galectins in tumors and in clinical immunotherapy. Int J Mol Sci 19, 430 (2018)PubMedCentral
179.
go back to reference G.J. Baker, P. Chockley, V.N. Yadav, R. Doherty, M. Ritt, S. Sivaramakrishnan, M.G. Castro, P.R. Lowenstein, Natural killer cells eradicate galectin-1-deficient glioma in the absence of adaptive immunity. Cancer Res 74, 5079–5090 (2014)PubMedPubMedCentral G.J. Baker, P. Chockley, V.N. Yadav, R. Doherty, M. Ritt, S. Sivaramakrishnan, M.G. Castro, P.R. Lowenstein, Natural killer cells eradicate galectin-1-deficient glioma in the absence of adaptive immunity. Cancer Res 74, 5079–5090 (2014)PubMedPubMedCentral
180.
go back to reference G.J. Baker, P. Chockley, D. Zamler, M.G. Castro, P.R. Lowenstein, Natural killer cells require monocytic gr-1(+)/CD11b(+) myeloid cells to eradicate orthotopically engrafted glioma cells. Oncoimmunol 5, e1163461 (2016) G.J. Baker, P. Chockley, D. Zamler, M.G. Castro, P.R. Lowenstein, Natural killer cells require monocytic gr-1(+)/CD11b(+) myeloid cells to eradicate orthotopically engrafted glioma cells. Oncoimmunol 5, e1163461 (2016)
181.
go back to reference J. Park, S.H. Wrzesinski, E. Stern, M. Look, J. Criscione, R. Ragheb, S.M. Jay, S.L. Demento, A. Agawu, P. Licona Limon, A.F. Ferrandino, D. Gonzalez, A. Habermann, R.A. Flavell, T.M. Fahmy, Combination delivery of TGF-β inhibitor and IL-2 by nanoscale liposomal polymeric gels enhances tumour immunotherapy. Nat Mater 11, 895–905 (2012)PubMedPubMedCentral J. Park, S.H. Wrzesinski, E. Stern, M. Look, J. Criscione, R. Ragheb, S.M. Jay, S.L. Demento, A. Agawu, P. Licona Limon, A.F. Ferrandino, D. Gonzalez, A. Habermann, R.A. Flavell, T.M. Fahmy, Combination delivery of TGF-β inhibitor and IL-2 by nanoscale liposomal polymeric gels enhances tumour immunotherapy. Nat Mater 11, 895–905 (2012)PubMedPubMedCentral
182.
go back to reference F. Otegbeye, E. Ojo, S. Moreton, N. Mackowski, D.A. Lee, M. de Lima, D.N. Wald, Inhibiting TGF-beta signaling preserves the function of highly activated, in vitro expanded natural killer cells in AML and colon cancer models. PLoS One 13, e0191358 (2018)PubMedPubMedCentral F. Otegbeye, E. Ojo, S. Moreton, N. Mackowski, D.A. Lee, M. de Lima, D.N. Wald, Inhibiting TGF-beta signaling preserves the function of highly activated, in vitro expanded natural killer cells in AML and colon cancer models. PLoS One 13, e0191358 (2018)PubMedPubMedCentral
183.
go back to reference A. Young, S.F. Ngiow, Y. Gao, A.M. Patch, D.S. Barkauskas, M. Messaoudene, G. Lin, J.D. Coudert, K.A. Stannard, L. Zitvogel, A2AR adenosine signaling suppresses natural killer cell maturation in the tumor microenvironment. Cancer Res 78, 1003–1016 (2018)PubMed A. Young, S.F. Ngiow, Y. Gao, A.M. Patch, D.S. Barkauskas, M. Messaoudene, G. Lin, J.D. Coudert, K.A. Stannard, L. Zitvogel, A2AR adenosine signaling suppresses natural killer cell maturation in the tumor microenvironment. Cancer Res 78, 1003–1016 (2018)PubMed
184.
go back to reference J. Wang, K.B. Lupo, A.M. Chambers, S. Matosevic, Purinergic targeting enhances immunotherapy of CD73+ solid tumors with piggyBac-engineered chimeric antigen receptor natural killer cells. J Immunother Cancer 6, 136 (2018)PubMedPubMedCentral J. Wang, K.B. Lupo, A.M. Chambers, S. Matosevic, Purinergic targeting enhances immunotherapy of CD73+ solid tumors with piggyBac-engineered chimeric antigen receptor natural killer cells. J Immunother Cancer 6, 136 (2018)PubMedPubMedCentral
185.
go back to reference O.E. Franco, A.K. Shaw, D.W. Strand, S.W. Hayward, Cancer associated fibroblasts in cancer pathogenesis. Semin Cell Dev Biol 21, 33–39 (2010)PubMed O.E. Franco, A.K. Shaw, D.W. Strand, S.W. Hayward, Cancer associated fibroblasts in cancer pathogenesis. Semin Cell Dev Biol 21, 33–39 (2010)PubMed
186.
go back to reference M. Augsten, Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front Oncol 4, 62 (2014)PubMedPubMedCentral M. Augsten, Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front Oncol 4, 62 (2014)PubMedPubMedCentral
187.
go back to reference G. Valcz, F. Sipos, Z. Tulassay, B. Molnar, Y. Yagi, Importance of carcinoma-associated fibroblast-derived proteins in clinical oncology. J Clin Pathol 67, 1026–1031 (2014)PubMed G. Valcz, F. Sipos, Z. Tulassay, B. Molnar, Y. Yagi, Importance of carcinoma-associated fibroblast-derived proteins in clinical oncology. J Clin Pathol 67, 1026–1031 (2014)PubMed
188.
go back to reference T. Li, S. Yi, W. Liu, C. Jia, G. Wang, X. Hua, Y. Tai, Q. Zhang, G. Chen, Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Med Oncol 30, 663 (2013)PubMed T. Li, S. Yi, W. Liu, C. Jia, G. Wang, X. Hua, Y. Tai, Q. Zhang, G. Chen, Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Med Oncol 30, 663 (2013)PubMed
189.
go back to reference M. Balsamo, F. Scordamaglia, G. Pietra, C. Manzini, C. Cantoni, M. Boitano, P. Queirolo, W. Vermi, F. Facchetti, A. Moretta, L. Moretta, M.C. Mingari, M. Vitale, Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proc Natl Acad Sci U S A 106, 20847–20852 (2009)PubMedPubMedCentral M. Balsamo, F. Scordamaglia, G. Pietra, C. Manzini, C. Cantoni, M. Boitano, P. Queirolo, W. Vermi, F. Facchetti, A. Moretta, L. Moretta, M.C. Mingari, M. Vitale, Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proc Natl Acad Sci U S A 106, 20847–20852 (2009)PubMedPubMedCentral
190.
go back to reference T. Inoue, K. Adachi, K. Kawana, A. Taguchi, T. Nagamatsu, A. Fujimoto, K. Tomio, A. Yamashita, S. Eguchi, H. Nishida, H. Nakamura, M. Sato, M. Yoshida, T. Arimoto, O. Wada-Hiraike, K. Oda, Y. Osuga, T. Fujii, Cancer-associated fibroblast suppresses killing activity of natural killer cells through downregulation of poliovirus receptor (PVR/CD155), a ligand of activating NK receptor. International J Oncol 49, 1297–1304 (2016) T. Inoue, K. Adachi, K. Kawana, A. Taguchi, T. Nagamatsu, A. Fujimoto, K. Tomio, A. Yamashita, S. Eguchi, H. Nishida, H. Nakamura, M. Sato, M. Yoshida, T. Arimoto, O. Wada-Hiraike, K. Oda, Y. Osuga, T. Fujii, Cancer-associated fibroblast suppresses killing activity of natural killer cells through downregulation of poliovirus receptor (PVR/CD155), a ligand of activating NK receptor. International J Oncol 49, 1297–1304 (2016)
191.
go back to reference R. Zhang, F. Qi, F. Zhao, G. Li, S. Shao, X. Zhang, L. Yuan, Y. Feng, Cancer-associated fibroblasts enhance tumor-associated macrophages enrichment and suppress NK cells function in colorectal cancer. Cell Death Dis 10, 273 (2019)PubMedPubMedCentral R. Zhang, F. Qi, F. Zhao, G. Li, S. Shao, X. Zhang, L. Yuan, Y. Feng, Cancer-associated fibroblasts enhance tumor-associated macrophages enrichment and suppress NK cells function in colorectal cancer. Cell Death Dis 10, 273 (2019)PubMedPubMedCentral
192.
go back to reference R. Francescone, D. B. Vendramini-Costa, J. Franco-Barraza, J. Wagner, A. Muir, A. N. Lau, L. Gabitova, T. Pazina, S. Gupta, T. Luong, N. Shah, D. Rollins, R. Malik, R. Thapa, D. Restifo, Y. Zhou, K. Q. Cai, H. H. Hensley, Y. Tan, W. D. Kruger, K. Devarajan, S. Balachandran, W. S. El-Deiry, M. G. V. Heiden, S. Peri, K. S. Campbell, I. Astsaturov, E. Cukierman. The NetrinG1/NGL-1 Axis promotes pancreatic tumorigenesis through cancer associated fibroblast driven nutritional support and immunosuppression. BioRxiv 330209 (2019) R. Francescone, D. B. Vendramini-Costa, J. Franco-Barraza, J. Wagner, A. Muir, A. N. Lau, L. Gabitova, T. Pazina, S. Gupta, T. Luong, N. Shah, D. Rollins, R. Malik, R. Thapa, D. Restifo, Y. Zhou, K. Q. Cai, H. H. Hensley, Y. Tan, W. D. Kruger, K. Devarajan, S. Balachandran, W. S. El-Deiry, M. G. V. Heiden, S. Peri, K. S. Campbell, I. Astsaturov, E. Cukierman. The NetrinG1/NGL-1 Axis promotes pancreatic tumorigenesis through cancer associated fibroblast driven nutritional support and immunosuppression. BioRxiv 330209 (2019)
193.
go back to reference M.H. Townsend, G. Shrestha, R.A. Robison, K.L. O’Neill, The expansion of targetable biomarkers for CAR T cell therapy. J Exp Clin Cancer Res 37, 163 (2018)PubMedPubMedCentral M.H. Townsend, G. Shrestha, R.A. Robison, K.L. O’Neill, The expansion of targetable biomarkers for CAR T cell therapy. J Exp Clin Cancer Res 37, 163 (2018)PubMedPubMedCentral
194.
go back to reference L.C.S. Wang, A. Lo, J. Scholler, J. Sun, R.S. Majumdar, V. Kapoor, M. Antzis, C.E. Cotner, L.A. Johnson, A.C. Durham, C.C. Solomides, C.H. June, E. Puré, S.M. Albelda, Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res 2, 154–166 (2014) L.C.S. Wang, A. Lo, J. Scholler, J. Sun, R.S. Majumdar, V. Kapoor, M. Antzis, C.E. Cotner, L.A. Johnson, A.C. Durham, C.C. Solomides, C.H. June, E. Puré, S.M. Albelda, Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res 2, 154–166 (2014)
195.
go back to reference Y.J. Xie, M. Dougan, N. Jailkhani, J. Ingram, T. Fang, L. Kummer, N. Momin, N. Pishesha, S. Rickelt, R.O. Hynes, H. Ploegh, Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc Natl Acad Sci U S A 116, 7624–7631 (2019)PubMedPubMedCentral Y.J. Xie, M. Dougan, N. Jailkhani, J. Ingram, T. Fang, L. Kummer, N. Momin, N. Pishesha, S. Rickelt, R.O. Hynes, H. Ploegh, Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc Natl Acad Sci U S A 116, 7624–7631 (2019)PubMedPubMedCentral
196.
go back to reference A. Lo, L.C.S. Wang, J. Scholler, J. Monslow, D. Avery, K. Newick, S. O’Brien, R.A. Evans, D.J. Bajor, C. Clendenin, A.C. Durham, E.L. Buza, R.H. Vonderheide, C.H. June, S.M. Albelda, E. Puré, Tumor-promoting Desmoplasia is disrupted by depleting FAP-expressing stromal cells. Cancer Res 75, 2800–2810 (2015)PubMedPubMedCentral A. Lo, L.C.S. Wang, J. Scholler, J. Monslow, D. Avery, K. Newick, S. O’Brien, R.A. Evans, D.J. Bajor, C. Clendenin, A.C. Durham, E.L. Buza, R.H. Vonderheide, C.H. June, S.M. Albelda, E. Puré, Tumor-promoting Desmoplasia is disrupted by depleting FAP-expressing stromal cells. Cancer Res 75, 2800–2810 (2015)PubMedPubMedCentral
197.
go back to reference A. O’Connell, S. Wang, L.M. Weiner, Abstract A096: The potential role of fibroblast activation protein as a natural killer cell immune checkpoint. Cancer Immunol Res 7, A096–A096 (2019) A. O’Connell, S. Wang, L.M. Weiner, Abstract A096: The potential role of fibroblast activation protein as a natural killer cell immune checkpoint. Cancer Immunol Res 7, A096–A096 (2019)
198.
go back to reference C. Hutmacher, N. Núñez, A.R. Liuzzi, B. Becher, D. Neri, Targeted delivery of IL2 to the tumor stroma potentiates the action of immune checkpoint inhibitors by preferential activation of NK and CD8+ T cells. Cancer Immunol. Res 7, 572–583 (2019) C. Hutmacher, N. Núñez, A.R. Liuzzi, B. Becher, D. Neri, Targeted delivery of IL2 to the tumor stroma potentiates the action of immune checkpoint inhibitors by preferential activation of NK and CD8+ T cells. Cancer Immunol. Res 7, 572–583 (2019)
199.
go back to reference M. Moschetta, F. Pretto, A. Berndt, K. Galler, P. Richter, A. Bassi, P. Oliva, E. Micotti, G. Valbusa, K. Schwager, M. Kaspar, E. Trachsel, H. Kosmehl, M.R. Bani, D. Neri, R. Giavazzi, Paclitaxel enhances therapeutic efficacy of the F8-IL2 Immunocytokine to EDA-fibronectin–positive metastatic human melanoma xenografts. Cancer Res 72, 1814–1824 (2012)PubMed M. Moschetta, F. Pretto, A. Berndt, K. Galler, P. Richter, A. Bassi, P. Oliva, E. Micotti, G. Valbusa, K. Schwager, M. Kaspar, E. Trachsel, H. Kosmehl, M.R. Bani, D. Neri, R. Giavazzi, Paclitaxel enhances therapeutic efficacy of the F8-IL2 Immunocytokine to EDA-fibronectin–positive metastatic human melanoma xenografts. Cancer Res 72, 1814–1824 (2012)PubMed
200.
go back to reference J. Tchou, Y. Zhao, B.L. Levine, P.J. Zhang, M.M. Davis, J.J. Melenhorst, I. Kulikovskaya, A.L. Brennan, X. Liu, S.F. Lacey, A.D. Posey, A.D. Williams, A. So, J.R. Conejo-Garcia, G. Plesa, R.M. Young, S. McGettigan, J. Campbell, R.H. Pierce, J.M. Matro, A.M. DeMichele, A.S. Clark, L.J. Cooper, L.M. Schuchter, R.H. Vonderheide, C.H. June, Safety and efficacy of intratumoral injections of chimeric antigen receptor (CAR) T cells in metastatic breast cancer. Cancer Immunol Res 5, 1152–1161 (2017) J. Tchou, Y. Zhao, B.L. Levine, P.J. Zhang, M.M. Davis, J.J. Melenhorst, I. Kulikovskaya, A.L. Brennan, X. Liu, S.F. Lacey, A.D. Posey, A.D. Williams, A. So, J.R. Conejo-Garcia, G. Plesa, R.M. Young, S. McGettigan, J. Campbell, R.H. Pierce, J.M. Matro, A.M. DeMichele, A.S. Clark, L.J. Cooper, L.M. Schuchter, R.H. Vonderheide, C.H. June, Safety and efficacy of intratumoral injections of chimeric antigen receptor (CAR) T cells in metastatic breast cancer. Cancer Immunol Res 5, 1152–1161 (2017)
201.
go back to reference A. Klampatsa, D.Y. Achkova, D.M. Davies, A.C. Parente-Pereira, N. Woodman, J. Rosekilly, G. Osborne, T. Thayaparan, A. Bille, M. Sheaf, J.F. Spicer, J. King, J. Maher, Intracavitary “T4 immunotherapy” of malignant mesothelioma using pan-ErbB re-targeted CAR T-cells. Cancer Lett 393, 52–59 (2017)PubMed A. Klampatsa, D.Y. Achkova, D.M. Davies, A.C. Parente-Pereira, N. Woodman, J. Rosekilly, G. Osborne, T. Thayaparan, A. Bille, M. Sheaf, J.F. Spicer, J. King, J. Maher, Intracavitary “T4 immunotherapy” of malignant mesothelioma using pan-ErbB re-targeted CAR T-cells. Cancer Lett 393, 52–59 (2017)PubMed
202.
go back to reference S.J. Priceman, D. Tilakawardane, B. Jeang, B. Aguilar, J.P. Murad, A.K. Park, W.C. Chang, J.R. Ostberg, J. Neman, R. Jandial, J. Portnow, S.J. Forman, C.E. Brown, Regional delivery of chimeric antigen receptor-engineered T cells effectively targets HER2+ breast Cancer metastasis to the brain. Clin Cancer Res 24, 95–105 (2018)PubMed S.J. Priceman, D. Tilakawardane, B. Jeang, B. Aguilar, J.P. Murad, A.K. Park, W.C. Chang, J.R. Ostberg, J. Neman, R. Jandial, J. Portnow, S.J. Forman, C.E. Brown, Regional delivery of chimeric antigen receptor-engineered T cells effectively targets HER2+ breast Cancer metastasis to the brain. Clin Cancer Res 24, 95–105 (2018)PubMed
203.
go back to reference A. Nellan, C. Rota, R. Majzner, C.M. Lester-McCully, A.M. Griesinger, J.M. Mulcahy Levy, N.K. Foreman, K.E. Warren, D.W. Lee, Durable regression of Medulloblastoma after regional and intravenous delivery of anti-HER2 chimeric antigen receptor T cells. J Immunother Cancer 6, 30 (2018)PubMedPubMedCentral A. Nellan, C. Rota, R. Majzner, C.M. Lester-McCully, A.M. Griesinger, J.M. Mulcahy Levy, N.K. Foreman, K.E. Warren, D.W. Lee, Durable regression of Medulloblastoma after regional and intravenous delivery of anti-HER2 chimeric antigen receptor T cells. J Immunother Cancer 6, 30 (2018)PubMedPubMedCentral
204.
go back to reference S.C. Katz, G.R. Point, M. Cunetta, M. Thorn, P. Guha, N.J. Espat, C. Boutros, N. Hanna, R.P. Junghans, Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery. Cancer Gene Ther 23, 142–148 (2016)PubMedPubMedCentral S.C. Katz, G.R. Point, M. Cunetta, M. Thorn, P. Guha, N.J. Espat, C. Boutros, N. Hanna, R.P. Junghans, Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery. Cancer Gene Ther 23, 142–148 (2016)PubMedPubMedCentral
205.
go back to reference S. Genßler, M.C. Burger, C. Zhang, S. Oelsner, I. Mildenberger, M. Wagner, J.P. Steinbach, W.S. Wels, Dual targeting of glioblastoma with chimeric antigen receptor-engineered natural killer cells overcomes heterogeneity of target antigen expression and enhances antitumor activity and survival. Oncoimmunol 5, e1119354 (2016) S. Genßler, M.C. Burger, C. Zhang, S. Oelsner, I. Mildenberger, M. Wagner, J.P. Steinbach, W.S. Wels, Dual targeting of glioblastoma with chimeric antigen receptor-engineered natural killer cells overcomes heterogeneity of target antigen expression and enhances antitumor activity and survival. Oncoimmunol 5, e1119354 (2016)
206.
go back to reference C. Zhang, M.C. Burger, L. Jennewein, S. Genßler, K. Schönfeld, P. Zeiner, E. Hattingen, P.N. Harter, M. Mittelbronn, T. Tonn, J.P. Steinbach, W.S. Wels, ErbB2/HER2-specific NK cells for targeted therapy of glioblastoma. J Natl Cancer Inst 108(5) (2016) C. Zhang, M.C. Burger, L. Jennewein, S. Genßler, K. Schönfeld, P. Zeiner, E. Hattingen, P.N. Harter, M. Mittelbronn, T. Tonn, J.P. Steinbach, W.S. Wels, ErbB2/HER2-specific NK cells for targeted therapy of glioblastoma. J Natl Cancer Inst 108(5) (2016)
207.
go back to reference J. Han, J. Chu, W. Keung Chan, J. Zhang, Y. Wang, J.B. Cohen, A. Victor, W.H. Meisen, S.H. Kim, P. Grandi, Q.E. Wang, X. He, I. Nakano, E.A. Chiocca, J.C. Glorioso Iii, B. Kaur, M.A. Caligiuri, J. Yu, CAR-engineered NK cells targeting wild-type EGFR and EGFRvIII enhance killing of glioblastoma and patient-derived glioblastoma stem cells. Sci Rep 5, 11483 (2016) J. Han, J. Chu, W. Keung Chan, J. Zhang, Y. Wang, J.B. Cohen, A. Victor, W.H. Meisen, S.H. Kim, P. Grandi, Q.E. Wang, X. He, I. Nakano, E.A. Chiocca, J.C. Glorioso Iii, B. Kaur, M.A. Caligiuri, J. Yu, CAR-engineered NK cells targeting wild-type EGFR and EGFRvIII enhance killing of glioblastoma and patient-derived glioblastoma stem cells. Sci Rep 5, 11483 (2016)
208.
go back to reference X. Chen, J. Han, J. Chu, L. Zhang, J. Zhang, C. Chen, L. Chen, Y. Wang, H. Wang, L. Yi, J.B. Elder, Q.E. Wang, X. He, B. Kaur, E.A. Chiocca, J. Yu, A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 7, 27764–27777 (2016)PubMedPubMedCentral X. Chen, J. Han, J. Chu, L. Zhang, J. Zhang, C. Chen, L. Chen, Y. Wang, H. Wang, L. Yi, J.B. Elder, Q.E. Wang, X. He, B. Kaur, E.A. Chiocca, J. Yu, A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 7, 27764–27777 (2016)PubMedPubMedCentral
209.
go back to reference V. Bachanova, S. Cooley, T.E. Defor, M.R. Verneris, B. Zhang, D.H. McKenna, J. Curtsinger, A. Panoskaltsis-Mortari, D. Lewis, K. Hippen, P. McGlave, D.J. Weisdorf, B.R. Blazar, J.S. Miller, Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood 123, 3855–3863 (2014) V. Bachanova, S. Cooley, T.E. Defor, M.R. Verneris, B. Zhang, D.H. McKenna, J. Curtsinger, A. Panoskaltsis-Mortari, D. Lewis, K. Hippen, P. McGlave, D.J. Weisdorf, B.R. Blazar, J.S. Miller, Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood 123, 3855–3863 (2014)
210.
go back to reference E. Liu, Y. Tong, G. Dotti, H. Shaim, B. Savoldo, M. Mukherjee, J. Orange, X. Wan, X. Lu, A. Reynolds, M. Gagea, P. Banerjee, R. Cai, M.H. Bdaiwi, R. Basar, M. Muftuoglu, L. Li, D. Marin, W. Wierda, M. Keating, R. Champlin, E. Shpall, K. Rezvani, Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 32, 520–531 (2017)PubMedPubMedCentral E. Liu, Y. Tong, G. Dotti, H. Shaim, B. Savoldo, M. Mukherjee, J. Orange, X. Wan, X. Lu, A. Reynolds, M. Gagea, P. Banerjee, R. Cai, M.H. Bdaiwi, R. Basar, M. Muftuoglu, L. Li, D. Marin, W. Wierda, M. Keating, R. Champlin, E. Shpall, K. Rezvani, Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 32, 520–531 (2017)PubMedPubMedCentral
211.
go back to reference P.W. Stacpoole, Therapeutic targeting of the pyruvate dehydrogenase complex/pyruvate dehydrogenase kinase (PDC/PDK) Axis in Cancer. J Natl Cancer Inst 109, 11 (2017) P.W. Stacpoole, Therapeutic targeting of the pyruvate dehydrogenase complex/pyruvate dehydrogenase kinase (PDC/PDK) Axis in Cancer. J Natl Cancer Inst 109, 11 (2017)
212.
go back to reference N. Assmann, K.L. O’Brien, R.P. Donnelly, L. Dyck, V. Zaiatz-Bittencourt, R.M. Loftus, P. Heinrich, P.J. Oefner, L. Lynch, C.M. Gardiner, K. Dettmer, D.K. Finlay, Srebp-controlled glucose metabolism is essential for NK cell functional responses. Nat Immunol 18, 1197–1206 (2017)PubMed N. Assmann, K.L. O’Brien, R.P. Donnelly, L. Dyck, V. Zaiatz-Bittencourt, R.M. Loftus, P. Heinrich, P.J. Oefner, L. Lynch, C.M. Gardiner, K. Dettmer, D.K. Finlay, Srebp-controlled glucose metabolism is essential for NK cell functional responses. Nat Immunol 18, 1197–1206 (2017)PubMed
213.
go back to reference M. Sukumar, J. Liu, Y. Ji, M. Subramanian, J.G. Crompton, Z. Yu, R. Roychoudhuri, D.C. Palmer, P. Muranski, E.D. Karoly, R.P. Mohney, C.A. Klebanoff, A. Lal, T. Finkel, N.P. Restifo, L. Gattinoni, Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J Clin Invest 123, 4479–4488 (2013)PubMedPubMedCentral M. Sukumar, J. Liu, Y. Ji, M. Subramanian, J.G. Crompton, Z. Yu, R. Roychoudhuri, D.C. Palmer, P. Muranski, E.D. Karoly, R.P. Mohney, C.A. Klebanoff, A. Lal, T. Finkel, N.P. Restifo, L. Gattinoni, Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J Clin Invest 123, 4479–4488 (2013)PubMedPubMedCentral
214.
go back to reference J.G. Crompton, M. Sukumar, R. Roychoudhuri, D. Clever, A. Gros, R.L. Eil, E. Tran, K.I. Hanada, Z. Yu, D.C. Palmer, S.P. Kerkar, R.D. Michalek, T. Upham, A. Leonardi, N. Acquavella, E. Wang, F.M. Marincola, L. Gattinoni, P. Muranski, M.S. Sundrud, C.A. Klebanoff, S.A. Rosenberg, D.T. Fearon, N.P. Restifo, Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics. Cancer Res 75, 296–305 (2015)PubMed J.G. Crompton, M. Sukumar, R. Roychoudhuri, D. Clever, A. Gros, R.L. Eil, E. Tran, K.I. Hanada, Z. Yu, D.C. Palmer, S.P. Kerkar, R.D. Michalek, T. Upham, A. Leonardi, N. Acquavella, E. Wang, F.M. Marincola, L. Gattinoni, P. Muranski, M.S. Sundrud, C.A. Klebanoff, S.A. Rosenberg, D.T. Fearon, N.P. Restifo, Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics. Cancer Res 75, 296–305 (2015)PubMed
215.
go back to reference O.U. Kawalekar, R.S. O’Connor, J.A. Fraietta, L. Guo, S.E. McGettigan, A.D. Posey, P.R. Patel, S. Guedan, J. Scholler, B. Keith, N.W. Snyder, I.A. Blair, M.C. Milone, C.H. June, Distinct signaling of Coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. 44, 380–390 (2016)PubMed O.U. Kawalekar, R.S. O’Connor, J.A. Fraietta, L. Guo, S.E. McGettigan, A.D. Posey, P.R. Patel, S. Guedan, J. Scholler, B. Keith, N.W. Snyder, I.A. Blair, M.C. Milone, C.H. June, Distinct signaling of Coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. 44, 380–390 (2016)PubMed
216.
go back to reference E.M. McWilliams, J.M. Mele, C. Cheney, E.A. Timmerman, F. Fiazuddin, E.J. Strattan, X. Mo, J.C. Byrd, N. Muthusamy, F.T. Awan, Therapeutic CD94/NKG2A blockade improves natural killer cell dysfunction in chronic lymphocytic leukemia. Oncoimmunol 5, e1226720 (2016) E.M. McWilliams, J.M. Mele, C. Cheney, E.A. Timmerman, F. Fiazuddin, E.J. Strattan, X. Mo, J.C. Byrd, N. Muthusamy, F.T. Awan, Therapeutic CD94/NKG2A blockade improves natural killer cell dysfunction in chronic lymphocytic leukemia. Oncoimmunol 5, e1226720 (2016)
217.
go back to reference T.N. Dao, S. Matosevic, Immunometabolic responses of natural killer cells to inhibitory tumor microenvironment checkpoints. Immunometabolism 1, e20190003 (2019) T.N. Dao, S. Matosevic, Immunometabolic responses of natural killer cells to inhibitory tumor microenvironment checkpoints. Immunometabolism 1, e20190003 (2019)
218.
go back to reference M. Shevtsov, G. Multhoff, Immunological and translational aspects of NK cell-based antitumor immunotherapies. Front Immunol 7, 492 (2016)PubMedPubMedCentral M. Shevtsov, G. Multhoff, Immunological and translational aspects of NK cell-based antitumor immunotherapies. Front Immunol 7, 492 (2016)PubMedPubMedCentral
220.
go back to reference M. Turcotte, K. Spring, S. Pommey, G. Chouinard, I. Cousineau, J. George, G.M. Chen, D.M. Gendoo, B. Haibe-Kains, T. Karn, CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res 75, 4494–4503 (2015)PubMed M. Turcotte, K. Spring, S. Pommey, G. Chouinard, I. Cousineau, J. George, G.M. Chen, D.M. Gendoo, B. Haibe-Kains, T. Karn, CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res 75, 4494–4503 (2015)PubMed
221.
go back to reference Z. Gao, H. Wang, F. Lin, X. Wang, M. Long, H. Zhang, K. Dong, CD73 promotes proliferation and migration of human cervical cancer cells independent of its enzyme activity. BMC Cancer 17, 135 (2017)PubMedPubMedCentral Z. Gao, H. Wang, F. Lin, X. Wang, M. Long, H. Zhang, K. Dong, CD73 promotes proliferation and migration of human cervical cancer cells independent of its enzyme activity. BMC Cancer 17, 135 (2017)PubMedPubMedCentral
222.
go back to reference S. Loi, S. Pommey, B. Haibe-Kains, P.A. Beavis, P.K. Darcy, M.J. Smyth, J. Stagg, CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc Natl Acad Sci U S A 110, 11091–11096 (2013)PubMedPubMedCentral S. Loi, S. Pommey, B. Haibe-Kains, P.A. Beavis, P.K. Darcy, M.J. Smyth, J. Stagg, CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc Natl Acad Sci U S A 110, 11091–11096 (2013)PubMedPubMedCentral
223.
go back to reference M. Lupia, F. Angiolini, G. Bertalot, S. Freddi, K.F. Sachsenmeier, E. Chisci, B. Kutryb-Zajac, S. Confalonieri, R.T. Smolenski, R. Giovannoni, CD73 regulates Stemness and epithelial-mesenchymal transition in ovarian Cancer-initiating cells. Stem Cell Rep 10, 1412–1425 (2018) M. Lupia, F. Angiolini, G. Bertalot, S. Freddi, K.F. Sachsenmeier, E. Chisci, B. Kutryb-Zajac, S. Confalonieri, R.T. Smolenski, R. Giovannoni, CD73 regulates Stemness and epithelial-mesenchymal transition in ovarian Cancer-initiating cells. Stem Cell Rep 10, 1412–1425 (2018)
224.
go back to reference L. Antonioli, C. Blandizzi, F. Malavasi, D. Ferrari, G. Haskó, Anti-CD73 immunotherapy: A viable way to reprogram the tumor microenvironment. Oncoimmunol 5, e1216292 (2016) L. Antonioli, C. Blandizzi, F. Malavasi, D. Ferrari, G. Haskó, Anti-CD73 immunotherapy: A viable way to reprogram the tumor microenvironment. Oncoimmunol 5, e1216292 (2016)
225.
go back to reference J. C. Geoghegan, G. Diedrich, X. Lu, K. Rosenthal, K. F. Sachsenmeier, H. Wu, W. F. Dall’Acqua, M. M. Damschroder, Inhibition of CD73 AMP hhydrolysis by a therapeutic antibody with a dual, non-competitive mechanism of action. MAbs 8, 454–467 (2016) J. C. Geoghegan, G. Diedrich, X. Lu, K. Rosenthal, K. F. Sachsenmeier, H. Wu, W. F. Dall’Acqua, M. M. Damschroder, Inhibition of CD73 AMP hhydrolysis by a therapeutic antibody with a dual, non-competitive mechanism of action. MAbs 8, 454–467 (2016)
226.
go back to reference I. Perrot, H.A. Michaud, M. Giraudon-Paoli, S. Augier, A. Docquier, L. Gros, R. Courtois, C. Déjou, D. Jecko, O. Becquart, H. Rispaud-Blanc, L. Gauthier, B. Rossi, S. Chanteux, N. Gourdin, B. Amigues, A. Roussel, A. Bensussan, J.F. Eliaou, J. Bastid, F. Romagné, Y. Morel, E. Narni-Mancinelli, E. Vivier, C. Paturel, N. Bonnefoy, Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies. Cell Rep 27, 2411–2425 (2019) I. Perrot, H.A. Michaud, M. Giraudon-Paoli, S. Augier, A. Docquier, L. Gros, R. Courtois, C. Déjou, D. Jecko, O. Becquart, H. Rispaud-Blanc, L. Gauthier, B. Rossi, S. Chanteux, N. Gourdin, B. Amigues, A. Roussel, A. Bensussan, J.F. Eliaou, J. Bastid, F. Romagné, Y. Morel, E. Narni-Mancinelli, E. Vivier, C. Paturel, N. Bonnefoy, Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies. Cell Rep 27, 2411–2425 (2019)
227.
go back to reference H.R. Lee, C.H. Son, E.K. Koh, J.H. Bae, C.D. Kang, K. Yang, Y.S. Park, Expansion of cytotoxic natural killer cells using irradiated autologous peripheral blood mononuclear cells and anti-CD16 antibody. Sci Rep 7, 11075 (2017)PubMedPubMedCentral H.R. Lee, C.H. Son, E.K. Koh, J.H. Bae, C.D. Kang, K. Yang, Y.S. Park, Expansion of cytotoxic natural killer cells using irradiated autologous peripheral blood mononuclear cells and anti-CD16 antibody. Sci Rep 7, 11075 (2017)PubMedPubMedCentral
228.
go back to reference S. Kumar, Natural killer cell cytotoxicity and its regulation by inhibitory receptors. Immunology 154, 383–393 (2018) S. Kumar, Natural killer cell cytotoxicity and its regulation by inhibitory receptors. Immunology 154, 383–393 (2018)
229.
go back to reference M. Sabry, A. Zubiak, S.P. Hood, P. Simmonds, H. Arellano-Ballestero, E. Cournoyer, M. Mashar, A.G. Pockley, M.W. Lowdell, Tumor- and cytokine-primed human natural killer cells exhibit distinct phenotypic and transcriptional signatures. PLoS One 14, e0218674 (2019)PubMedPubMedCentral M. Sabry, A. Zubiak, S.P. Hood, P. Simmonds, H. Arellano-Ballestero, E. Cournoyer, M. Mashar, A.G. Pockley, M.W. Lowdell, Tumor- and cytokine-primed human natural killer cells exhibit distinct phenotypic and transcriptional signatures. PLoS One 14, e0218674 (2019)PubMedPubMedCentral
230.
go back to reference M. Felices, T.R. Lenvik, Z.B. Davis, J.S. Miller, D.A. Vallera, Generation of BiKEs and TriKEs to improve NK cell-mediated targeting of tumor cells. Methods Mol Biol 1441, 333–346 (2016)PubMedPubMedCentral M. Felices, T.R. Lenvik, Z.B. Davis, J.S. Miller, D.A. Vallera, Generation of BiKEs and TriKEs to improve NK cell-mediated targeting of tumor cells. Methods Mol Biol 1441, 333–346 (2016)PubMedPubMedCentral
231.
go back to reference J.U. Schmohl, M. Felices, E. Taras, J.S. Miller, D.A. Vallera, Enhanced ADCC and NK cell activation of an Anticarcinoma bispecific antibody by genetic insertion of a modified IL-15 cross-linker. Mol Ther 24, 1312–1322 (2016)PubMedPubMedCentral J.U. Schmohl, M. Felices, E. Taras, J.S. Miller, D.A. Vallera, Enhanced ADCC and NK cell activation of an Anticarcinoma bispecific antibody by genetic insertion of a modified IL-15 cross-linker. Mol Ther 24, 1312–1322 (2016)PubMedPubMedCentral
232.
go back to reference D.A. Vallera, M. Felices, R. McElmurry, V. McCullar, X. Zhou, J.U. Schmohl, B. Zhang, A.J. Lenvik, A. Panoskaltsis-Mortari, M.R. Verneris, J. Tolar, S. Cooley, D.J. Weisdorf, B.R. Blazar, J.S. Miller, IL15 Trispecific killer engagers (TriKE) make natural killer cells specific to CD33+ targets while also inducing persistence, in vivo expansion, and enhanced function. Clin Cancer Res 22, 3440–3450 (2016)PubMedPubMedCentral D.A. Vallera, M. Felices, R. McElmurry, V. McCullar, X. Zhou, J.U. Schmohl, B. Zhang, A.J. Lenvik, A. Panoskaltsis-Mortari, M.R. Verneris, J. Tolar, S. Cooley, D.J. Weisdorf, B.R. Blazar, J.S. Miller, IL15 Trispecific killer engagers (TriKE) make natural killer cells specific to CD33+ targets while also inducing persistence, in vivo expansion, and enhanced function. Clin Cancer Res 22, 3440–3450 (2016)PubMedPubMedCentral
233.
go back to reference L. Gauthier, A. Morel, N. Anceriz, B. Rossi, A. Blanchard-Alvarez, G. Grondin, S. Trichard, C. Cesari, M. Sapet, F. Bosco, H. Rispaud-Blanc, F. Guillot, S. Cornen, A. Roussel, B. Amigues, G. Habif, F. Caraguel, S. Arrufat, R. Remark, F. Romagné, Y. Morel, E. Narni-Mancinelli, E. Vivier, Multifunctional natural killer cell engagers targeting NKp46 trigger protective tumor immunity. Cell 177, 1701–1713 (2019) L. Gauthier, A. Morel, N. Anceriz, B. Rossi, A. Blanchard-Alvarez, G. Grondin, S. Trichard, C. Cesari, M. Sapet, F. Bosco, H. Rispaud-Blanc, F. Guillot, S. Cornen, A. Roussel, B. Amigues, G. Habif, F. Caraguel, S. Arrufat, R. Remark, F. Romagné, Y. Morel, E. Narni-Mancinelli, E. Vivier, Multifunctional natural killer cell engagers targeting NKp46 trigger protective tumor immunity. Cell 177, 1701–1713 (2019)
234.
go back to reference X. Yuan, H. Wu, H. Bu, J. Zhou, H. Zhang, Targeting the immunity protein kinases for immuno-oncology. Eur J Med Chem 163, 413–427 (2019)PubMed X. Yuan, H. Wu, H. Bu, J. Zhou, H. Zhang, Targeting the immunity protein kinases for immuno-oncology. Eur J Med Chem 163, 413–427 (2019)PubMed
235.
go back to reference J.M. Yingling, W.T. McMillen, L. Yan, H. Huang, J.S. Sawyer, J. Graff, D.K. Clawson, K.S. Britt, B.D. Anderson, D.W. Beight, D. Desaiah, M.M. Lahn, K.A. Benhadji, M.J. Lallena, R.B. Holmgaard, X. Xu, F. Zhang, J.R. Manro, P.W. Iversen, C.V. Iyer, R.A. Brekken, M.D. Kalos, K.E. Driscoll, Preclinical assessment of galunisertib (LY2157299 monohydrate), a first-in-class transforming growth factor-β receptor type I inhibitor. Oncotarget 9, 6659–6677 (2018)PubMed J.M. Yingling, W.T. McMillen, L. Yan, H. Huang, J.S. Sawyer, J. Graff, D.K. Clawson, K.S. Britt, B.D. Anderson, D.W. Beight, D. Desaiah, M.M. Lahn, K.A. Benhadji, M.J. Lallena, R.B. Holmgaard, X. Xu, F. Zhang, J.R. Manro, P.W. Iversen, C.V. Iyer, R.A. Brekken, M.D. Kalos, K.E. Driscoll, Preclinical assessment of galunisertib (LY2157299 monohydrate), a first-in-class transforming growth factor-β receptor type I inhibitor. Oncotarget 9, 6659–6677 (2018)PubMed
236.
go back to reference H.C. Tran, Z. Wan, M.A. Sheard, J. Sun, J.R. Jackson, J. Malvar, Y. Xu, L. Wang, R. Sposto, E.S. Kim, S. Asgharzadeh, R.C. Seeger, TGFβR1 Blockade with Galunisertib (LY2157299) Enhances Anti-Neuroblastoma Activity of the Anti-GD2 Antibody Dinutuximab (ch14.18) with Natural Killer Cells. Clin. Cancer Res 23, 804–813 (2017)PubMed H.C. Tran, Z. Wan, M.A. Sheard, J. Sun, J.R. Jackson, J. Malvar, Y. Xu, L. Wang, R. Sposto, E.S. Kim, S. Asgharzadeh, R.C. Seeger, TGFβR1 Blockade with Galunisertib (LY2157299) Enhances Anti-Neuroblastoma Activity of the Anti-GD2 Antibody Dinutuximab (ch14.18) with Natural Killer Cells. Clin. Cancer Res 23, 804–813 (2017)PubMed
237.
go back to reference D. Vijayan, A. Young, M.W. Teng, M.J. Smyth, Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer 17, 709–724 (2017)PubMed D. Vijayan, A. Young, M.W. Teng, M.J. Smyth, Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer 17, 709–724 (2017)PubMed
238.
go back to reference R.D. Leone, L.A. Emens, Targeting adenosine for cancer immunotherapy. J Immunother Cancer 6, 57 (2018) R.D. Leone, L.A. Emens, Targeting adenosine for cancer immunotherapy. J Immunother Cancer 6, 57 (2018)
239.
go back to reference R.D. Leone, Y.C. Lo, J.D. Powell, A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy. Comp Struct Biotechnol J 13, 265–272 (2015) R.D. Leone, Y.C. Lo, J.D. Powell, A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy. Comp Struct Biotechnol J 13, 265–272 (2015)
240.
go back to reference M. Congreve, G.A. Brown, A. Borodovsky, M.L. Lamb, Targeting adenosine A2A receptor antagonism for treatment of cancer. Expert Opin Drug Discov 13, 997–1003 (2018)PubMed M. Congreve, G.A. Brown, A. Borodovsky, M.L. Lamb, Targeting adenosine A2A receptor antagonism for treatment of cancer. Expert Opin Drug Discov 13, 997–1003 (2018)PubMed
241.
go back to reference K. Sek, C. Mølck, G.D. Stewart, L. Kats, P.K. Darcy, P.A. Beavis, Targeting adenosine receptor signaling in Cancer immunotherapy. Int J Mol Sci 19, 12 (2018) K. Sek, C. Mølck, G.D. Stewart, L. Kats, P.K. Darcy, P.A. Beavis, Targeting adenosine receptor signaling in Cancer immunotherapy. Int J Mol Sci 19, 12 (2018)
242.
go back to reference P.A. Beavis, U. Divisekera, C. Paget, M.T. Chow, L.B. John, C. Devaud, K. Dwyer, J. Stagg, M.J. Smyth, P.K. Darcy, Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci U S A 110, 14711–14716 (2013)PubMedPubMedCentral P.A. Beavis, U. Divisekera, C. Paget, M.T. Chow, L.B. John, C. Devaud, K. Dwyer, J. Stagg, M.J. Smyth, P.K. Darcy, Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci U S A 110, 14711–14716 (2013)PubMedPubMedCentral
243.
go back to reference A. Young, S.F. Ngiow, J. Madore, J. Reinhardt, J. Landsberg, A. Chitsazan, J. Rautela, T. Bald, D.S. Barkauskas, E. Ahern, N.D. Huntington, D. Schadendorf, G.V. Long, G.M. Boyle, M. Hölzel, R.A. Scolyer, M.J. Smyth, Targeting adenosine in BRAF-mutant melanoma reduces tumor growth and metastasis. Cancer Res 77, 4684–4696 (2017)PubMed A. Young, S.F. Ngiow, J. Madore, J. Reinhardt, J. Landsberg, A. Chitsazan, J. Rautela, T. Bald, D.S. Barkauskas, E. Ahern, N.D. Huntington, D. Schadendorf, G.V. Long, G.M. Boyle, M. Hölzel, R.A. Scolyer, M.J. Smyth, Targeting adenosine in BRAF-mutant melanoma reduces tumor growth and metastasis. Cancer Res 77, 4684–4696 (2017)PubMed
244.
go back to reference R. Romee, J.W. Leong, T.A. Fehniger, Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer. Scientifica 2014, 205796 (2014)PubMedPubMedCentral R. Romee, J.W. Leong, T.A. Fehniger, Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer. Scientifica 2014, 205796 (2014)PubMedPubMedCentral
245.
go back to reference N.D. Huntington, H. Puthalakath, P. Gunn, E. Naik, E.M. Michalak, M.J. Smyth, H. Tabarias, M.A. Degli-Esposti, G. Dewson, S.N. Willis, N. Motoyama, D.C.S. Huang, S.L. Nutt, D.M. Tarlinton, A. Strasser, Interleukin 15-mediated survival of natural killer cells is determined by interactions among Bim, Noxa and Mcl-1. Nat Immunol 8, 856–863 (2007)PubMedPubMedCentral N.D. Huntington, H. Puthalakath, P. Gunn, E. Naik, E.M. Michalak, M.J. Smyth, H. Tabarias, M.A. Degli-Esposti, G. Dewson, S.N. Willis, N. Motoyama, D.C.S. Huang, S.L. Nutt, D.M. Tarlinton, A. Strasser, Interleukin 15-mediated survival of natural killer cells is determined by interactions among Bim, Noxa and Mcl-1. Nat Immunol 8, 856–863 (2007)PubMedPubMedCentral
246.
go back to reference E. Mortier, T. Woo, R. Advincula, S. Gozalo, A. Ma, IL-15Ralpha chaperones IL-15 to stable dendritic cell membrane complexes that activate NK cells via trans presentation. J Exp Med 205, 1213–1225 (2008)PubMedPubMedCentral E. Mortier, T. Woo, R. Advincula, S. Gozalo, A. Ma, IL-15Ralpha chaperones IL-15 to stable dendritic cell membrane complexes that activate NK cells via trans presentation. J Exp Med 205, 1213–1225 (2008)PubMedPubMedCentral
247.
go back to reference R. Evans, J.A. Fuller, G. Christianson, D.M. Krupke, A.B. Troutt, IL-15 mediates anti-tumor effects after cyclophosphamide injection of tumor-bearing mice and enhances adoptive immunotherapy: The potential role of NK cell subpopulations. Cell Immunol 179, 66–73 (1997)PubMed R. Evans, J.A. Fuller, G. Christianson, D.M. Krupke, A.B. Troutt, IL-15 mediates anti-tumor effects after cyclophosphamide injection of tumor-bearing mice and enhances adoptive immunotherapy: The potential role of NK cell subpopulations. Cell Immunol 179, 66–73 (1997)PubMed
248.
go back to reference A.H. Pillet, J. Thèze, T. Rose, Interleukin (IL)-2 and IL-15 have different effects on human natural killer lymphocytes. Hum Immunol 72, 1013–1017 (2011)PubMed A.H. Pillet, J. Thèze, T. Rose, Interleukin (IL)-2 and IL-15 have different effects on human natural killer lymphocytes. Hum Immunol 72, 1013–1017 (2011)PubMed
249.
go back to reference R.J. Hennessy, K. Pham, R. Delconte, J. Rautela, P.D. Hodgkin, N.D. Huntington, Quantifying NK cell growth and survival changes in response to cytokines and regulatory checkpoint blockade helps identify optimal culture and expansion conditions. J Leukoc Biol 105, 1341–1354 (2019)PubMed R.J. Hennessy, K. Pham, R. Delconte, J. Rautela, P.D. Hodgkin, N.D. Huntington, Quantifying NK cell growth and survival changes in response to cytokines and regulatory checkpoint blockade helps identify optimal culture and expansion conditions. J Leukoc Biol 105, 1341–1354 (2019)PubMed
250.
go back to reference M. Strengell, S. Matikainen, J. Sirén, A. Lehtonen, D. Foster, I. Julkunen, T. Sareneva, IL-21 in synergy with IL-15 or IL-18 enhances IFN-gamma production in human NK and T cells. J Immunol 170, 5464–5469 (2003)PubMed M. Strengell, S. Matikainen, J. Sirén, A. Lehtonen, D. Foster, I. Julkunen, T. Sareneva, IL-21 in synergy with IL-15 or IL-18 enhances IFN-gamma production in human NK and T cells. J Immunol 170, 5464–5469 (2003)PubMed
251.
go back to reference E. Lusty, S.M. Poznanski, K. Kwofie, T.S. Mandur, D.A. Lee, C.D. Richards, A.A. Ashkar, IL-18/IL-15/IL-12 synergy induces elevated and prolonged IFN-γ production by ex vivo expanded NK cells which is not due to enhanced STAT4 activation. Mol Immunol 88, 138–147 (2017)PubMed E. Lusty, S.M. Poznanski, K. Kwofie, T.S. Mandur, D.A. Lee, C.D. Richards, A.A. Ashkar, IL-18/IL-15/IL-12 synergy induces elevated and prolonged IFN-γ production by ex vivo expanded NK cells which is not due to enhanced STAT4 activation. Mol Immunol 88, 138–147 (2017)PubMed
252.
go back to reference C.S. Henney, K. Kuribayashi, D.E. Kern, S. Gillis, Interleukin-2 augments natural killer cell activity. Nature 291, 335–338 (1981) C.S. Henney, K. Kuribayashi, D.E. Kern, S. Gillis, Interleukin-2 augments natural killer cell activity. Nature 291, 335–338 (1981)
253.
go back to reference R. Spolski, P. Li, W.J. Leonard, Biology and regulation of IL-2: From molecular mechanisms to human therapy. Nat. Rev. Immunol. 18, 648–659 (2018)PubMed R. Spolski, P. Li, W.J. Leonard, Biology and regulation of IL-2: From molecular mechanisms to human therapy. Nat. Rev. Immunol. 18, 648–659 (2018)PubMed
254.
255.
go back to reference T. Jiang, C. Zhou, S. Ren, Role of IL-2 in cancer immunotherapy. Oncoimmunol 5, e1163462 (2016) T. Jiang, C. Zhou, S. Ren, Role of IL-2 in cancer immunotherapy. Oncoimmunol 5, e1163462 (2016)
256.
go back to reference H. Choudhry, N. Helmi, W.H. Abdulaal, M. Zeyadi, M.A. Zamzami, W. Wu, M.M. Mahmoud, M.K. Warsi, M. Rasool, M.S. Jamal, Prospects of IL-2 in Cancer immunotherapy. Biomed Res Int 2018, 9056173 (2018)PubMedPubMedCentral H. Choudhry, N. Helmi, W.H. Abdulaal, M. Zeyadi, M.A. Zamzami, W. Wu, M.M. Mahmoud, M.K. Warsi, M. Rasool, M.S. Jamal, Prospects of IL-2 in Cancer immunotherapy. Biomed Res Int 2018, 9056173 (2018)PubMedPubMedCentral
257.
go back to reference T. Chinen, A.K. Kannan, A.G. Levine, X. Fan, U. Klein, Y. Zheng, G. Gasteiger, Y. Feng, J.D. Fontenot, A.Y. Rudensky, An essential role for the IL-2 receptor in Treg cell function. Nat Immunol 17, 1322–1333 (2016)PubMedPubMedCentral T. Chinen, A.K. Kannan, A.G. Levine, X. Fan, U. Klein, Y. Zheng, G. Gasteiger, Y. Feng, J.D. Fontenot, A.Y. Rudensky, An essential role for the IL-2 receptor in Treg cell function. Nat Immunol 17, 1322–1333 (2016)PubMedPubMedCentral
258.
go back to reference D.H. Charych, U. Hoch, J.L. Langowski, S.R. Lee, M.K. Addepalli, P.B. Kirk, D. Sheng, X. Liu, P.W. Sims, L.A. VanderVeen, C.F. Ali, T.K. Chang, M. Konakova, R.L. Pena, R.S. Kanhere, Y.M. Kirksey, C. Ji, Y. Wang, J. Huang, T.D. Sweeney, S.S. Kantak, S.K. Doberstein, NKTR-214, an engineered cytokine with biased IL2 receptor binding, increased tumor exposure, and marked efficacy in mouse tumor models. Clin Cancer Res 22, 680–690 (2016)PubMed D.H. Charych, U. Hoch, J.L. Langowski, S.R. Lee, M.K. Addepalli, P.B. Kirk, D. Sheng, X. Liu, P.W. Sims, L.A. VanderVeen, C.F. Ali, T.K. Chang, M. Konakova, R.L. Pena, R.S. Kanhere, Y.M. Kirksey, C. Ji, Y. Wang, J. Huang, T.D. Sweeney, S.S. Kantak, S.K. Doberstein, NKTR-214, an engineered cytokine with biased IL2 receptor binding, increased tumor exposure, and marked efficacy in mouse tumor models. Clin Cancer Res 22, 680–690 (2016)PubMed
259.
go back to reference V. Bachanova, J.S. Miller, NK cells in therapy of cancer. Crit Rev Oncog 19, 133–141 (2014) V. Bachanova, J.S. Miller, NK cells in therapy of cancer. Crit Rev Oncog 19, 133–141 (2014)
260.
go back to reference Z.B. Davis, M. Felices, M.R. Verneris, J.S. Miller, Natural killer cell adoptive transfer therapy: Exploiting the first line of defense against cancer. Cancer J 21, 486–491 (2015) Z.B. Davis, M. Felices, M.R. Verneris, J.S. Miller, Natural killer cell adoptive transfer therapy: Exploiting the first line of defense against cancer. Cancer J 21, 486–491 (2015)
261.
go back to reference S. Matosevic, Viral and nonviral engineering of natural killer cells as emerging adoptive Cancer immunotherapies. J Immunol Res 2018, 4054815 (2018)PubMedPubMedCentral S. Matosevic, Viral and nonviral engineering of natural killer cells as emerging adoptive Cancer immunotherapies. J Immunol Res 2018, 4054815 (2018)PubMedPubMedCentral
262.
go back to reference Z. Wang, L. Guo, Y. Song, Y. Zhang, D. Lin, B. Hu, Y. Mei, D. Sandikin, H. Liu, Augmented anti-tumor activity of NK-92 cells expressing chimeric receptors of TGF-βR II and NKG2D. Cancer Immunol Immunother 66, 537–548 (2017)PubMed Z. Wang, L. Guo, Y. Song, Y. Zhang, D. Lin, B. Hu, Y. Mei, D. Sandikin, H. Liu, Augmented anti-tumor activity of NK-92 cells expressing chimeric receptors of TGF-βR II and NKG2D. Cancer Immunol Immunother 66, 537–548 (2017)PubMed
263.
go back to reference W. Hu, G. Wang, D. Huang, M. Sui, Y. Xu, Cancer immunotherapy based on natural killer cells: Current Progress and new opportunities. Front Immunol 10, 1205 (2019)PubMedPubMedCentral W. Hu, G. Wang, D. Huang, M. Sui, Y. Xu, Cancer immunotherapy based on natural killer cells: Current Progress and new opportunities. Front Immunol 10, 1205 (2019)PubMedPubMedCentral
264.
go back to reference Y. Li, D.L. Hermanson, B.S. Moriarity, D.S. Kaufman, Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell 23, 181–192 (2018)PubMedPubMedCentral Y. Li, D.L. Hermanson, B.S. Moriarity, D.S. Kaufman, Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell 23, 181–192 (2018)PubMedPubMedCentral
265.
go back to reference M. Naeimi Kararoudi, H. Dolatshad, P. Trikha, S.R.A. Hussain, E. Elmas, J.A. Foltz, J.E. Moseman, A. Thakkar, R.J. Nakkula, M. Lamb, N. Chakravarti, K.J. McLaughlin, D.A. Lee, Generation of Knock-out primary and expanded human NK cells using Cas9 ribonucleoproteins. J Vis Exp 136, e58237 (2018) M. Naeimi Kararoudi, H. Dolatshad, P. Trikha, S.R.A. Hussain, E. Elmas, J.A. Foltz, J.E. Moseman, A. Thakkar, R.J. Nakkula, M. Lamb, N. Chakravarti, K.J. McLaughlin, D.A. Lee, Generation of Knock-out primary and expanded human NK cells using Cas9 ribonucleoproteins. J Vis Exp 136, e58237 (2018)
266.
go back to reference Q.M. Wang, P.M.K. Tang, G.Y. Lian, C. Li, J. Li, X.R. Huang, K. F. To, H.Y. Lan, Enhanced Cancer immunotherapy with Smad3-silenced NK-92 cells. Cancer Immunol Res 6, 965–977 (2018) Q.M. Wang, P.M.K. Tang, G.Y. Lian, C. Li, J. Li, X.R. Huang, K. F. To, H.Y. Lan, Enhanced Cancer immunotherapy with Smad3-silenced NK-92 cells. Cancer Immunol Res 6, 965–977 (2018)
267.
go back to reference E. S. Yvon, R. Burga, A. Powell, C. R. Cruz, R. Fernandes, C. Barese, T. Nguyen, M. S. Abdel-Baki, C. M. Bollard, Cord blood natural killer cells expressing a dominant negative TGF-β receptor: Implications for adoptive immunotherapy for glioblastoma. Cytotherapy 19, 408–418 (n.d.)(107) E. S. Yvon, R. Burga, A. Powell, C. R. Cruz, R. Fernandes, C. Barese, T. Nguyen, M. S. Abdel-Baki, C. M. Bollard, Cord blood natural killer cells expressing a dominant negative TGF-β receptor: Implications for adoptive immunotherapy for glioblastoma. Cytotherapy 19, 408–418 (n.d.)(107)
268.
go back to reference R. Esser, T. Müller, D. Stefes, S. Kloess, D. Seidel, S.D. Gillies, C. Aperlo-Iffland, J.S. Huston, C. Uherek, K. Schönfeld, T. Tonn, N. Huebener, H.N. Lode, U. Koehl, W.S. Wels, NK cells engineered to express a GD2 -specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J Cell Mol Med 16, 569–581 (2012)PubMedPubMedCentral R. Esser, T. Müller, D. Stefes, S. Kloess, D. Seidel, S.D. Gillies, C. Aperlo-Iffland, J.S. Huston, C. Uherek, K. Schönfeld, T. Tonn, N. Huebener, H.N. Lode, U. Koehl, W.S. Wels, NK cells engineered to express a GD2 -specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J Cell Mol Med 16, 569–581 (2012)PubMedPubMedCentral
269.
go back to reference C. Sahm, K. Schönfeld, W.S. Wels, Expression of IL-15 in NK cells results in rapid enrichment and selective cytotoxicity of gene-modified effectors that carry a tumor-specific antigen receptor. Cancer Immunol Immunother 61, 1451–1461 (2012)PubMed C. Sahm, K. Schönfeld, W.S. Wels, Expression of IL-15 in NK cells results in rapid enrichment and selective cytotoxicity of gene-modified effectors that carry a tumor-specific antigen receptor. Cancer Immunol Immunother 61, 1451–1461 (2012)PubMed
270.
go back to reference Y.H. Chang, J. Connolly, N. Shimasaki, K. Mimura, K. Kono, D. Campana, A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res 73, 1777–1786 (2013)PubMed Y.H. Chang, J. Connolly, N. Shimasaki, K. Mimura, K. Kono, D. Campana, A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res 73, 1777–1786 (2013)PubMed
271.
go back to reference R. Parihar, C. Rivas, M. Huynh, B. Omer, N. Lapteva, L.S. Metelitsa, S.M. Gottschalk, C.M. Rooney, NK cells expressing a chimeric activating receptor eliminate MDSCs and rescue impaired CAR-T cell activity against solid tumors. Cancer Immunol Res 7, 363–375 (2019) R. Parihar, C. Rivas, M. Huynh, B. Omer, N. Lapteva, L.S. Metelitsa, S.M. Gottschalk, C.M. Rooney, NK cells expressing a chimeric activating receptor eliminate MDSCs and rescue impaired CAR-T cell activity against solid tumors. Cancer Immunol Res 7, 363–375 (2019)
272.
go back to reference K. Schönfeld, C. Sahm, C. Zhang, S. Naundorf, C. Brendel, M. Odendahl, P. Nowakowska, H. Bönig, U. Köhl, S. Kloess, S. Köhler, H. Holtgreve-Grez, A. Jauch, M. Schmidt, R. Schubert, K. Kühlcke, E. Seifried, H.G. Klingemann, M.A. Rieger, T. Tonn, M. Grez, W.S. Wels, Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor. Mol Ther 23, 330–338 (2015)PubMed K. Schönfeld, C. Sahm, C. Zhang, S. Naundorf, C. Brendel, M. Odendahl, P. Nowakowska, H. Bönig, U. Köhl, S. Kloess, S. Köhler, H. Holtgreve-Grez, A. Jauch, M. Schmidt, R. Schubert, K. Kühlcke, E. Seifried, H.G. Klingemann, M.A. Rieger, T. Tonn, M. Grez, W.S. Wels, Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor. Mol Ther 23, 330–338 (2015)PubMed
273.
go back to reference Q. Zhang, K. Tian, J. Xu, H. Zhang, L. Li, Q. Fu, D. Chai, H. Li, J. Zheng, Synergistic effects of Cabozantinib and EGFR-specific CAR-NK-92 cells in renal cell carcinoma. J Immunol Res 2017, 6915912 (2017)PubMedPubMedCentral Q. Zhang, K. Tian, J. Xu, H. Zhang, L. Li, Q. Fu, D. Chai, H. Li, J. Zheng, Synergistic effects of Cabozantinib and EGFR-specific CAR-NK-92 cells in renal cell carcinoma. J Immunol Res 2017, 6915912 (2017)PubMedPubMedCentral
274.
go back to reference M. Yu, H. Luo, M. Fan, X. Wu, B. Shi, S. Di, Y. Liu, Z. Pan, H. Jiang, Z. Li, Development of GPC3-specific chimeric antigen receptor-engineered natural killer cells for the treatment of hepatocellular carcinoma. Mol Ther 26, 366–378 (2018)PubMed M. Yu, H. Luo, M. Fan, X. Wu, B. Shi, S. Di, Y. Liu, Z. Pan, H. Jiang, Z. Li, Development of GPC3-specific chimeric antigen receptor-engineered natural killer cells for the treatment of hepatocellular carcinoma. Mol Ther 26, 366–378 (2018)PubMed
275.
go back to reference H. Park, A. Awasthi, J. Ayello, Y. Chu, S. Riddell, J. Rosenblum, D.A. Lee, M.S. Cairo, ROR1-specific chimeric antigen receptor (CAR) NK cell immunotherapy for high risk neuroblastomas and sarcomas. Biol Blood Marrow Transplant 23, S136–S137 (2017) H. Park, A. Awasthi, J. Ayello, Y. Chu, S. Riddell, J. Rosenblum, D.A. Lee, M.S. Cairo, ROR1-specific chimeric antigen receptor (CAR) NK cell immunotherapy for high risk neuroblastomas and sarcomas. Biol Blood Marrow Transplant 23, S136–S137 (2017)
Metadata
Title
Functional and metabolic targeting of natural killer cells to solid tumors
Authors
Jiao Wang
Sandro Matosevic
Publication date
01-08-2020
Publisher
Springer Netherlands
Keyword
Solid Tumor
Published in
Cellular Oncology / Issue 4/2020
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-020-00523-7

Other articles of this Issue 4/2020

Cellular Oncology 4/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine