Skip to main content
Erschienen in: Cancer Immunology, Immunotherapy 7/2015

01.07.2015 | Focussed Research Review

Immune checkpoint combinations from mouse to man

verfasst von: Midan Ai, Michael A. Curran

Erschienen in: Cancer Immunology, Immunotherapy | Ausgabe 7/2015

Einloggen, um Zugang zu erhalten

Abstract

The discovery that antibody blockade of the T cell co-inhibitory receptor cytotoxic T lymphocyte-associated protein 4 (CTLA-4) can restore tumor immunity against many murine transplantable tumors leading to complete rejection of established cancer forever changed the field of immunotherapy. In more robust murine models as well as human cancer, however, CTLA-4 blockade alone can slow tumor growth and extend patient survival, but is rarely curative. Subsequent studies have revealed a large family of T cell immune checkpoint receptors which tumors engage to shield themselves from host immunity. As with CTLA-4, blockade of one of these additional inhibitory receptors, programmed death 1, has led to remarkable therapeutic responses against tumors of multiple lineages. Checkpoint monotherapy has demonstrated that durable, immune-mediated cures of established metastatic cancers are possible, yet the percentage of patients experiencing these outcomes remains low due to both redundant mechanisms of immune suppression in the tumor and limiting toxicity associated with some therapies. Thus, extending the curative potential of immunotherapy to a larger percentage of patients with a broader spectrum of malignancies will likely require combinations of co-inhibitory blockade and co-stimulatory activation designed to peel back multiple layers of tumor immune suppression while at the same time minimizing immune-mediated toxicity. As over a dozen T cell immune checkpoints and an additional dozen more co-stimulatory receptors have now been described, the challenge before us is to identify the most advantageous combinations of these agents based on the knowledge of their underlying biology and preclinical studies in murine tumor models.
Literatur
1.
Zurück zum Zitat Ehrlich P (1909) Über den jetzigen Stand der Chemotherapie. Ber Dtsch Chem Ges 42:17–47CrossRef Ehrlich P (1909) Über den jetzigen Stand der Chemotherapie. Ber Dtsch Chem Ges 42:17–47CrossRef
2.
Zurück zum Zitat Zinkernagel RM, Doherty PC (1974) Immunological surveillance against altered self components by sensitised T lymphocytes in lymphocytic choriomeningitis. Nature 251:547–548PubMedCrossRef Zinkernagel RM, Doherty PC (1974) Immunological surveillance against altered self components by sensitised T lymphocytes in lymphocytic choriomeningitis. Nature 251:547–548PubMedCrossRef
4.
Zurück zum Zitat Wick M, Dubey P, Koeppen H, Siegel CT, Fields PE, Chen L, Bluestone JA, Schreiber H (1997) Antigenic cancer cells grow progressively in immune hosts without evidence for T cell exhaustion or systemic anergy. J Exp Med 186:229–238PubMedCentralPubMedCrossRef Wick M, Dubey P, Koeppen H, Siegel CT, Fields PE, Chen L, Bluestone JA, Schreiber H (1997) Antigenic cancer cells grow progressively in immune hosts without evidence for T cell exhaustion or systemic anergy. J Exp Med 186:229–238PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Krummel MF, Allison JP (1995) CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 182:459–465PubMedCrossRef Krummel MF, Allison JP (1995) CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 182:459–465PubMedCrossRef
6.
Zurück zum Zitat Leach DR, Krummel MF, Allison JP (1996) Enhancement of antitumor immunity by CTLA-4 blockade. Science 271:1734–1736PubMedCrossRef Leach DR, Krummel MF, Allison JP (1996) Enhancement of antitumor immunity by CTLA-4 blockade. Science 271:1734–1736PubMedCrossRef
7.
Zurück zum Zitat Ishida Y, Agata Y, Shibahara K, Honjo T (1992) Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 11:3887–3895PubMedCentralPubMed Ishida Y, Agata Y, Shibahara K, Honjo T (1992) Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 11:3887–3895PubMedCentralPubMed
8.
Zurück zum Zitat Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellstrom KE, Mittler RS, Chen L (1997) Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med 3:682–685PubMedCrossRef Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellstrom KE, Mittler RS, Chen L (1997) Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med 3:682–685PubMedCrossRef
9.
Zurück zum Zitat Weinberg AD, Rivera MM, Prell R et al (2000) Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol 164:2160–2169PubMedCrossRef Weinberg AD, Rivera MM, Prell R et al (2000) Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol 164:2160–2169PubMedCrossRef
10.
Zurück zum Zitat Moran AE, Kovacsovics-Bankowski M, Weinberg AD (2013) The TNFRs OX40, 4-1BB, and CD40 as targets for cancer immunotherapy. Curr Opin Immunol 25:230–237PubMedCrossRef Moran AE, Kovacsovics-Bankowski M, Weinberg AD (2013) The TNFRs OX40, 4-1BB, and CD40 as targets for cancer immunotherapy. Curr Opin Immunol 25:230–237PubMedCrossRef
13.
Zurück zum Zitat Hutloff A, Dittrich AM, Beier KC, Eljaschewitsch B, Kraft R, Anagnostopoulos I, Kroczek RA (1999) ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 397:263–266PubMedCrossRef Hutloff A, Dittrich AM, Beier KC, Eljaschewitsch B, Kraft R, Anagnostopoulos I, Kroczek RA (1999) ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 397:263–266PubMedCrossRef
14.
Zurück zum Zitat Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, Thompson CB, Griesser H, Mak TW (1995) Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 270:985–988PubMedCrossRef Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, Thompson CB, Griesser H, Mak TW (1995) Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 270:985–988PubMedCrossRef
15.
Zurück zum Zitat Nishimura H, Nose M, Hiai H, Minato N, Honjo T (1999) Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 11:141–151PubMedCrossRef Nishimura H, Nose M, Hiai H, Minato N, Honjo T (1999) Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 11:141–151PubMedCrossRef
16.
Zurück zum Zitat Nishimura H, Okazaki T, Tanaka Y et al (2001) Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 291:319–322PubMedCrossRef Nishimura H, Okazaki T, Tanaka Y et al (2001) Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 291:319–322PubMedCrossRef
17.
Zurück zum Zitat Curran MA, Montalvo W, Yagita H, Allison JP (2010) PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A 107:4275–4280PubMedCentralPubMedCrossRef Curran MA, Montalvo W, Yagita H, Allison JP (2010) PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A 107:4275–4280PubMedCentralPubMedCrossRef
18.
Zurück zum Zitat Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ (2007) Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 27:111–122PubMedCentralPubMedCrossRef Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ (2007) Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 27:111–122PubMedCentralPubMedCrossRef
19.
Zurück zum Zitat Melero I, Grimaldi AM, Perez-Gracia JL, Ascierto PA (2013) Clinical development of immunostimulatory monoclonal antibodies and opportunities for combination. Clin Cancer Res 19:997–1008PubMedCrossRef Melero I, Grimaldi AM, Perez-Gracia JL, Ascierto PA (2013) Clinical development of immunostimulatory monoclonal antibodies and opportunities for combination. Clin Cancer Res 19:997–1008PubMedCrossRef
20.
Zurück zum Zitat Wolchok JD, Kluger H, Callahan MK et al (2013) Nivolumab plus ipilimumab in advanced melanoma. New Engl J Med 369:122–133PubMedCrossRef Wolchok JD, Kluger H, Callahan MK et al (2013) Nivolumab plus ipilimumab in advanced melanoma. New Engl J Med 369:122–133PubMedCrossRef
21.
Zurück zum Zitat Sznol M, Kluger HM, Callahan MK et al (2014) Survival, response duration, and activity by BRAF mutation (MT) status of nivolumab (NIVO, anti-PD-1, BMS-936558, ONO-4538) and ipilimumab (IPI) concurrent therapy in advanced melanoma (MEL). J Clin Oncol 32(5s Suppl):LBA9003 Sznol M, Kluger HM, Callahan MK et al (2014) Survival, response duration, and activity by BRAF mutation (MT) status of nivolumab (NIVO, anti-PD-1, BMS-936558, ONO-4538) and ipilimumab (IPI) concurrent therapy in advanced melanoma (MEL). J Clin Oncol 32(5s Suppl):LBA9003
22.
Zurück zum Zitat Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G (2013) Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res 73:3591–3603PubMedCentralPubMedCrossRef Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G (2013) Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res 73:3591–3603PubMedCentralPubMedCrossRef
23.
Zurück zum Zitat Callahan MK, Horak CE, Curran MA et al (2013) Peripheral and tumor immune correlates in patients with advanced melanoma treated with combination nivolumab (anti-PD-1, BMS-936558, ONO-4538) and ipilimumab. J Clin Oncol 31(Suppl):3003 Callahan MK, Horak CE, Curran MA et al (2013) Peripheral and tumor immune correlates in patients with advanced melanoma treated with combination nivolumab (anti-PD-1, BMS-936558, ONO-4538) and ipilimumab. J Clin Oncol 31(Suppl):3003
24.
Zurück zum Zitat Wang W, Yu D, Sarnaik AA, Yu B, Hall M, Morelli D, Zhang Y, Zhao X, Weber JS (2012) Biomarkers on melanoma patient T cells associated with ipilimumab treatment. J Transl Med 10:146PubMedCentralPubMedCrossRef Wang W, Yu D, Sarnaik AA, Yu B, Hall M, Morelli D, Zhang Y, Zhao X, Weber JS (2012) Biomarkers on melanoma patient T cells associated with ipilimumab treatment. J Transl Med 10:146PubMedCentralPubMedCrossRef
25.
Zurück zum Zitat Ng Tang D, Shen Y, Sun J, Wen S, Wolchok JD, Yuan J, Allison JP, Sharma P (2013) Increased frequency of ICOS+ CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol Res 1:229–234PubMedCrossRef Ng Tang D, Shen Y, Sun J, Wen S, Wolchok JD, Yuan J, Allison JP, Sharma P (2013) Increased frequency of ICOS+ CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol Res 1:229–234PubMedCrossRef
26.
Zurück zum Zitat Carthon BC, Wolchok JD, Yuan J et al (2010) Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin Cancer Res 16:2861–2871PubMedCentralPubMedCrossRef Carthon BC, Wolchok JD, Yuan J et al (2010) Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin Cancer Res 16:2861–2871PubMedCentralPubMedCrossRef
27.
Zurück zum Zitat Fu T, He Q, Sharma P (2011) The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res 71:5445–5454PubMedCrossRef Fu T, He Q, Sharma P (2011) The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res 71:5445–5454PubMedCrossRef
28.
Zurück zum Zitat Fan X, Quezada SA, Sepulveda MA, Sharma P, Allison JP (2014) Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J Exp Med 211:715–725PubMedCentralPubMedCrossRef Fan X, Quezada SA, Sepulveda MA, Sharma P, Allison JP (2014) Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J Exp Med 211:715–725PubMedCentralPubMedCrossRef
29.
Zurück zum Zitat Lee SW, Croft M (2009) 4-1BB as a therapeutic target for human disease. Adv Exp Med Biol 647:120–129PubMedCrossRef Lee SW, Croft M (2009) 4-1BB as a therapeutic target for human disease. Adv Exp Med Biol 647:120–129PubMedCrossRef
30.
Zurück zum Zitat Wang C, Lin GH, McPherson AJ, Watts TH (2009) Immune regulation by 4-1BB and 4-1BBL: complexities and challenges. Immunol Rev 229:192–215PubMedCrossRef Wang C, Lin GH, McPherson AJ, Watts TH (2009) Immune regulation by 4-1BB and 4-1BBL: complexities and challenges. Immunol Rev 229:192–215PubMedCrossRef
31.
Zurück zum Zitat Li B, Lin J, Vanroey M, Jure-Kunkel M, Jooss K (2007) Established B16 tumors are rejected following treatment with GM-CSF-secreting tumor cell immunotherapy in combination with anti-4-1BB mAb. Clin Immunol 125:76–87PubMedCrossRef Li B, Lin J, Vanroey M, Jure-Kunkel M, Jooss K (2007) Established B16 tumors are rejected following treatment with GM-CSF-secreting tumor cell immunotherapy in combination with anti-4-1BB mAb. Clin Immunol 125:76–87PubMedCrossRef
32.
Zurück zum Zitat Curran MA, Geiger TL, Montalvo W, Kim M, Reiner SL, Al-Shamkhani A, Sun JC, Allison JP (2013) Systemic 4-1BB activation induces a novel T cell phenotype driven by high expression of Eomesodermin. J Exp Med 210:743–755PubMedCentralPubMedCrossRef Curran MA, Geiger TL, Montalvo W, Kim M, Reiner SL, Al-Shamkhani A, Sun JC, Allison JP (2013) Systemic 4-1BB activation induces a novel T cell phenotype driven by high expression of Eomesodermin. J Exp Med 210:743–755PubMedCentralPubMedCrossRef
33.
Zurück zum Zitat Belcaid Z, Phallen JA, Zeng J et al (2014) Focal radiation therapy combined with 4-1BB activation and CTLA-4 blockade yields long-term survival and a protective antigen-specific memory response in a murine glioma model. PLoS One 9:e101764PubMedCentralPubMedCrossRef Belcaid Z, Phallen JA, Zeng J et al (2014) Focal radiation therapy combined with 4-1BB activation and CTLA-4 blockade yields long-term survival and a protective antigen-specific memory response in a murine glioma model. PLoS One 9:e101764PubMedCentralPubMedCrossRef
34.
Zurück zum Zitat Curran MA, Kim M, Montalvo W, Al-Shamkhani A, Allison JP (2011) Combination CTLA-4 blockade and 4-1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. PLoS One 6:e19499PubMedCentralPubMedCrossRef Curran MA, Kim M, Montalvo W, Al-Shamkhani A, Allison JP (2011) Combination CTLA-4 blockade and 4-1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. PLoS One 6:e19499PubMedCentralPubMedCrossRef
35.
Zurück zum Zitat Kocak E, Lute K, Chang X et al (2006) Combination therapy with anti-CTL antigen-4 and anti-4-1BB antibodies enhances cancer immunity and reduces autoimmunity. Cancer Res 66:7276–7284PubMedCrossRef Kocak E, Lute K, Chang X et al (2006) Combination therapy with anti-CTL antigen-4 and anti-4-1BB antibodies enhances cancer immunity and reduces autoimmunity. Cancer Res 66:7276–7284PubMedCrossRef
36.
Zurück zum Zitat Sun Y, Lin X, Chen HM, Wu Q, Subudhi SK, Chen L, Fu YX (2002) Administration of agonistic anti-4-1BB monoclonal antibody leads to the amelioration of experimental autoimmune encephalomyelitis. J Immunol 168:1457–1465PubMedCrossRef Sun Y, Lin X, Chen HM, Wu Q, Subudhi SK, Chen L, Fu YX (2002) Administration of agonistic anti-4-1BB monoclonal antibody leads to the amelioration of experimental autoimmune encephalomyelitis. J Immunol 168:1457–1465PubMedCrossRef
37.
Zurück zum Zitat Kim YH, Choi BK, Shin SM et al (2011) 4-1BB triggering ameliorates experimental autoimmune encephalomyelitis by modulating the balance between Th17 and regulatory T cells. J Immunol 187:1120–1128PubMedCrossRef Kim YH, Choi BK, Shin SM et al (2011) 4-1BB triggering ameliorates experimental autoimmune encephalomyelitis by modulating the balance between Th17 and regulatory T cells. J Immunol 187:1120–1128PubMedCrossRef
38.
Zurück zum Zitat Callahan MK, Yang A, Tandon S et al (2011) Evaluation of serum IL-17 levels during ipilimumab therapy: correlation with colitis. J Clin Oncol 29(Suppl):2505 Callahan MK, Yang A, Tandon S et al (2011) Evaluation of serum IL-17 levels during ipilimumab therapy: correlation with colitis. J Clin Oncol 29(Suppl):2505
39.
Zurück zum Zitat Vudattu NK, Waldron-Lynch F, Truman LA, Deng S, Preston-Hurlburt P, Torres R, Raycroft MT, Mamula MJ, Herold KC (2014) Humanized mice as a model for aberrant responses in human T cell immunotherapy. J Immunol 193:587–596PubMedCrossRef Vudattu NK, Waldron-Lynch F, Truman LA, Deng S, Preston-Hurlburt P, Torres R, Raycroft MT, Mamula MJ, Herold KC (2014) Humanized mice as a model for aberrant responses in human T cell immunotherapy. J Immunol 193:587–596PubMedCrossRef
40.
Zurück zum Zitat Ascierto PA, Simeone E, Sznol M, Fu YX, Melero I (2010) Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. Semin Oncol 37:508–516PubMedCrossRef Ascierto PA, Simeone E, Sznol M, Fu YX, Melero I (2010) Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. Semin Oncol 37:508–516PubMedCrossRef
41.
Zurück zum Zitat Kvistborg P, Philips D, Kelderman S et al (2014) Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med 6:254ra128PubMedCrossRef Kvistborg P, Philips D, Kelderman S et al (2014) Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med 6:254ra128PubMedCrossRef
42.
Zurück zum Zitat Spranger S, Bao R, Gajewski TF (2014) Melanoma-intrinsic β-catenin signaling prevents T cell infiltration and anti-tumor immunity. J ImmunoTherapy Cancer 2(Suppl 3):O15CrossRef Spranger S, Bao R, Gajewski TF (2014) Melanoma-intrinsic β-catenin signaling prevents T cell infiltration and anti-tumor immunity. J ImmunoTherapy Cancer 2(Suppl 3):O15CrossRef
43.
Zurück zum Zitat Gubin MM, Zhang X, Schuster H et al (2014) Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515:577–581PubMedCentralPubMedCrossRef Gubin MM, Zhang X, Schuster H et al (2014) Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515:577–581PubMedCentralPubMedCrossRef
44.
Zurück zum Zitat van Rooij N, van Buuren MM, Philips D et al (2013) Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 31:e439–e442PubMedCrossRef van Rooij N, van Buuren MM, Philips D et al (2013) Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 31:e439–e442PubMedCrossRef
Metadaten
Titel
Immune checkpoint combinations from mouse to man
verfasst von
Midan Ai
Michael A. Curran
Publikationsdatum
01.07.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Immunology, Immunotherapy / Ausgabe 7/2015
Print ISSN: 0340-7004
Elektronische ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-014-1650-8

Weitere Artikel der Ausgabe 7/2015

Cancer Immunology, Immunotherapy 7/2015 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.