Skip to main content

Advertisement

Log in

Tumor-induced escape mechanisms and their association with resistance to checkpoint inhibitor therapy

  • Focussed Research Review
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

Immunotherapy aims to activate the immune system to fight cancer in a very specific and targeted manner. Despite the success of different immunotherapeutic strategies, in particular antibodies directed against checkpoints as well as adoptive T-cell therapy, the response of patients is limited in different types of cancers. This attributes to escape of the tumor from immune surveillance and development of acquired resistances during therapy. In this review, the different evasion and resistance mechanisms that limit the efficacy of immunotherapies targeting tumor-associated antigens presented by major histocompatibility complex molecules on the surface of the malignant cells are summarized. Overcoming these escape mechanisms is a great challenge, but might lead to a better clinical outcome of patients and is therefore currently a major focus of research.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Abbreviations

DAC:

5-Aza-2′-desoxycytidine

3′ UTR:

3′ Untranslated region

ACT:

Adoptive cell therapy

APM:

Antigen processing and presentation machinery

BGN:

Biglycan

CTL:

Cytotoxic T lymphocyte

CTLA-4:

Cytotoxic T-lymphocyte associated protein-4

DCN:

Decorin

ER:

Endoplasmic reticulum

ERAP:

Endoplasmic reticulum aminopeptidase

EBV:

Epstein–Barr virus

GAS:

Gamma activated site

HC:

Heavy chain

HNRNPR:

Heterogeneous nuclear ribonucleoprotein R

HCMV:

Human cytomegalovirus

iCP:

Immune checkpoint

iCPI:

Immune checkpoint inhibitor

im-miRNAs:

Immune modulatory miRNAs

ILT:

Inhibitory receptor Ig-like transcript

IFN:

Interferon

IRF:

Interferon regulated factor

ISRE:

Interferon-sensitive response element

LOH:

Loss of heterozygosity

Luc:

Luciferase

MHC:

Major histocompatibility complex

β2-m:

β2-Microglobulin

miRNA:

MicroRNA

MSI:

Microsatellite instability

Mex:

Muscle excess

MDSC:

Myeloid-derived suppressor cells

NLRC5:

NOD-like receptor family and acid domain-containing protein 5

OS:

Overall survival

Treg:

Regulatory T cell(s)

RBP:

RNA-binding proteins

RNAseq:

RNA sequencing

STAT:

Signal transducer and activator of transcription

SNP:

Single nucleotide polymorphism

SLRP:

Small leucine-rich proteoglycan

TPN:

Tapasin

TCGA:

The Cancer Genome Atlas

TGF-β:

Transforming growth factor β

TAP:

Transporter associated with antigen processing

TAF:

Tumor-associated fibroblast(s)

TAM:

Tumor-associated macrophages

TAN:

Tumor-associated neutrophil(s)

TMB:

Tumor mutational burden

TME:

Tumor microenvironment

References

  1. Leone P et al (2013) MHC class I antigen processing and presenting machinery: organization, function, and defects in tumor cells. J Natl Cancer Inst 105(16):1172–1187

    CAS  PubMed  Google Scholar 

  2. Zanker D, Chen W (2014) Standard and immunoproteasomes show similar peptide degradation specificities. Eur J Immunol 44(12):3500–3503

    CAS  PubMed  Google Scholar 

  3. Saveanu L et al (2005) Concerted peptide trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic reticulum. Nat Immunol 6(7):689–697

    CAS  PubMed  Google Scholar 

  4. Norbury CC, Eisenlohr LC (2016) Editorial overview: antigen processing. Curr Opin Immunol 40:5–6

    Google Scholar 

  5. Mintern JD, Macri C, Villadangos JA (2015) Modulation of antigen presentation by intracellular trafficking. Curr Opin Immunol 34:16–21

    CAS  PubMed  Google Scholar 

  6. Han LY et al (2008) HLA class I antigen processing machinery component expression and intratumoral T-Cell infiltrate as independent prognostic markers in ovarian carcinoma. Clin Cancer Res 14(11):3372–3379

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Perea F et al (2017) The absence of HLA class I expression in non-small cell lung cancer correlates with the tumor tissue structure and the pattern of T cell infiltration. Int J Cancer 140(4):888–899

    CAS  PubMed  Google Scholar 

  8. Garrido F et al (2017) The escape of cancer from T cell-mediated immune surveillance: HLA class I loss and tumor tissue architecture. Vaccines (Basel) 5(1):7

    Google Scholar 

  9. Seliger B (2016) Role of microRNAs on HLA-G expression in human tumors. Hum Immunol 77(9):760–763

    CAS  PubMed  Google Scholar 

  10. Real LM et al (1999) Expression of HLA G in human tumors is not a frequent event. Int J Cancer 81(4):512–518

    CAS  PubMed  Google Scholar 

  11. Davies B et al (2001) HLA-G expression by tumors. Am J Reprod Immunol 45(2):103–107

    CAS  PubMed  Google Scholar 

  12. Polakova K et al (2003) Analysis of HLA-G expression in malignant hematopoietic cells from leukemia patients. Leuk Res 27(7):643–648

    CAS  PubMed  Google Scholar 

  13. Frumento G et al (2000) Melanomas and melanoma cell lines do not express HLA-G, and the expression cannot be induced by gamma IFN treatment. Tissue Antigens 56(1):30–37

    CAS  PubMed  Google Scholar 

  14. Pangault C et al (1999) HLA-G protein expression is not induced during malignant transformation. Tissue Antigens 53(4 Pt 1):335–346

    CAS  PubMed  Google Scholar 

  15. Chang CC, Ferrone S (2003) HLA-G in melanoma: can the current controversies be solved? Semin Cancer Biol 13(5):361–369

    CAS  PubMed  Google Scholar 

  16. Wang Y et al (2011) Expression of HLA-G in patients with hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 10(2):158–163

    CAS  PubMed  Google Scholar 

  17. Yie SM et al (2007) Expression of human leucocyte antigen G (HLA-G) is associated with prognosis in non-small cell lung cancer. Lung Cancer 58(2):267–274

    PubMed  Google Scholar 

  18. Tronik-Le Roux D et al (2017) Novel landscape of HLA-G isoforms expressed in clear cell renal cell carcinoma patients. Mol Oncol 11(11):1561–1578

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Lin A, Yan WH (2018) Heterogeneity of HLA-G expression in cancers: facing the challenges. Front Immunol 9:2164

    PubMed  PubMed Central  Google Scholar 

  20. Lin A, Yan WH (2015) Human leukocyte antigen-G (HLA-G) expression in cancers: roles in immune evasion, metastasis and target for therapy. Mol Med 21(1):782–791

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Contini P et al (2003) Soluble HLA-A,-B,-C and -G molecules induce apoptosis in T and NK CD8 + cells and inhibit cytotoxic T cell activity through CD8 ligation. Eur J Immunol 33(1):125–134

    CAS  PubMed  Google Scholar 

  22. Menier C et al (2002) MICA triggering signal for NK cell tumor lysis is counteracted by HLA-G1-mediated inhibitory signal. Int J Cancer 100(1):63–70

    CAS  PubMed  Google Scholar 

  23. Fons P et al (2006) Soluble HLA-G1 inhibits angiogenesis through an apoptotic pathway and by direct binding to CD160 receptor expressed by endothelial cells. Blood 108(8):2608–2615

    CAS  PubMed  Google Scholar 

  24. Morandi F et al (2010) A novel mechanism of soluble HLA-G mediated immune modulation: downregulation of T cell chemokine receptor expression and impairment of chemotaxis. PLoS One 5(7):e11763

    PubMed  PubMed Central  Google Scholar 

  25. Morandi F et al (2011) Soluble HLA-G dampens CD94/NKG2A expression and function and differentially modulates chemotaxis and cytokine and chemokine secretion in CD56bright and CD56dim NK cells. Blood 118(22):5840–5850

    CAS  PubMed  Google Scholar 

  26. Agaugue S, Carosella ED, Rouas-Freiss N (2011) Role of HLA-G in tumor escape through expansion of myeloid-derived suppressor cells and cytokinic balance in favor of Th2 versus Th1/Th17. Blood 117(26):7021–7031

    CAS  PubMed  Google Scholar 

  27. Loumagne L et al (2014) In vivo evidence that secretion of HLA-G by immunogenic tumor cells allows their evasion from immunosurveillance. Int J Cancer 135(9):2107–2117

    CAS  PubMed  Google Scholar 

  28. Seliger B (2014) The link between MHC class I abnormalities of tumors, oncogenes, tumor suppressor genes, and transcription factors. J Immunotoxicol 11(4):308–310

    CAS  PubMed  Google Scholar 

  29. Cai L et al (2018) Defective HLA class I antigen processing machinery in cancer. Cancer Immunol Immunother 67(6):999–1009

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Donia M et al (2017) Acquired Immune resistance follows complete tumor regression without loss of target antigens or IFN gamma signaling. Cancer Res 77(17):4562–4566

    CAS  PubMed  Google Scholar 

  31. Kloor M, Michel S, von Knebel Doeberitz M (2010) Immune evasion of microsatellite unstable colorectal cancers. Int J Cancer 127(5):1001–1010

    CAS  PubMed  Google Scholar 

  32. Hicklin DJ et al (1998) Beta2-Microglobulin mutations, HLA class I antigen loss, and tumor progression in melanoma. J Clin Invest 101(12):2720–2729

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Chang CC et al (2005) Immune selection of hot-spot beta 2-microglobulin gene mutations, HLA-A2 allospecificity loss, and antigen-processing machinery component down-regulation in melanoma cells derived from recurrent metastases following immunotherapy. J Immunol 174(3):1462–1471

    CAS  PubMed  Google Scholar 

  34. Geertsen R et al (2002) Loss of single HLA class I allospecificities in melanoma cells due to selective genomic abbreviations. Int J Cancer 99(1):82–87

    CAS  PubMed  Google Scholar 

  35. del Campo AB et al (2014) Immune escape of cancer cells with beta2-microglobulin loss over the course of metastatic melanoma. Int J Cancer 134(1):102–113

    PubMed  Google Scholar 

  36. McGranahan N et al (2017) Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171(6):1259–1271

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Zehir A et al (2017) Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 23(6):703–713

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Seliger B (2017) Immune modulatory microRNAs as a novel mechanism to revert immune escape of tumors. Cytokine Growth Factor Rev 36:49–56

    CAS  PubMed  Google Scholar 

  39. Ritter C et al (2017) Epigenetic priming restores the HLA class-I antigen processing machinery expression in Merkel cell carcinoma. Sci Rep 7(1):2290

    PubMed  PubMed Central  Google Scholar 

  40. Jongsma MLM, Guarda G, Spaapen RM (2017) The regulatory network behind MHC class I expression. Mol Immunol. https://doi.org/10.1016/j.molimm.2017.12.005

    Article  PubMed  Google Scholar 

  41. Drukker M et al (2002) Characterization of the expression of MHC proteins in human embryonic stem cells. Proc Natl Acad Sci USA 99(15):9864–9869

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Vlkova V et al (2014) Epigenetic regulations in the IFNgamma signalling pathway: IFNgamma-mediated MHC class I upregulation on tumour cells is associated with DNA demethylation of antigen-presenting machinery genes. Oncotarget 5(16):6923–6935

    PubMed  PubMed Central  Google Scholar 

  43. Manning J et al (2008) Induction of MHC class I molecule cell surface expression and epigenetic activation of antigen-processing machinery components in a murine model for human papilloma virus 16-associated tumours. Immunology 123(2):218–227

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Setiadi AF et al (2008) Epigenetic enhancement of antigen processing and presentation promotes immune recognition of tumors. Cancer Res 68(23):9601–9607

    CAS  PubMed  Google Scholar 

  45. Dunker K et al (2008) Expression and regulation of non-classical HLA-G in renal cell carcinoma. Tissue Antigens 72(2):137–148

    CAS  PubMed  Google Scholar 

  46. Ramsuran V et al (2015) Epigenetic regulation of differential HLA-A allelic expression levels. Hum Mol Genet 24(15):4268–4275

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Gustafsson JR et al (2018) DNMT1 regulates expression of MHC class I in post-mitotic neurons. Mol Brain 11(1):36

    PubMed  PubMed Central  Google Scholar 

  48. Khan AN, Gregorie CJ, Tomasi TB (2008) Histone deacetylase inhibitors induce TAP, LMP, Tapasin genes and MHC class I antigen presentation by melanoma cells. Cancer Immunol Immunother 57(5):647–654

    CAS  PubMed  Google Scholar 

  49. Komatsu Y, Hayashi H (1998) Histone deacetylase inhibitors up-regulate the expression of cell surface MHC class-I molecules in B16/BL6 cells. J Antibiot (Tokyo) 51(1):89–91

    CAS  Google Scholar 

  50. Robbins GR et al (2012) Regulation of class I major histocompatibility complex (MHC) by nucleotide-binding domain, leucine-rich repeat-containing (NLR) proteins. J Biol Chem 287(29):24294–24303

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Moreau P et al (2003) HLA-G gene repression is reversed by demethylation. Proc Natl Acad Sci USA 100(3):1191–1196

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Holling TM et al (2009) Genetic and epigenetic control of the major histocompatibility complex class Ib gene HLA-G in trophoblast cell lines. Ann N Y Acad Sci 1173:538–544

    CAS  PubMed  Google Scholar 

  53. van den Elsen PJ (2011) Expression regulation of major histocompatibility complex class I and class II encoding genes. Front Immunol 2:48

    PubMed  PubMed Central  Google Scholar 

  54. van den Elsen PJ et al (1998) Regulation of MHC class I and II gene transcription: differences and similarities. Immunogenetics 48(3):208–221

    PubMed  Google Scholar 

  55. Howcroft TK et al (2003) Distinct transcriptional pathways regulate basal and activated major histocompatibility complex class I expression. Mol Cell Biol 23(10):3377–3391

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Gobin SJ et al (1998) The role of enhancer A in the locus-specific transactivation of classical and nonclassical HLA class I genes by nuclear factor kappa B. J Immunol 161(5):2276–2283

    CAS  PubMed  Google Scholar 

  57. Gobin SJ et al (1999) Transactivation of classical and nonclassical HLA class I genes through the IFN-stimulated response element. J Immunol 163(3):1428–1434

    CAS  PubMed  Google Scholar 

  58. Gobin SJ et al (2001) The MHC-specific enhanceosome and its role in MHC class I and beta(2)-microglobulin gene transactivation. J Immunol 167(9):5175–5184

    CAS  PubMed  Google Scholar 

  59. van den Elsen PJ et al (2004) Transcriptional regulation of antigen presentation. Curr Opin Immunol 16(1):67–75

    PubMed  Google Scholar 

  60. Gobin SJ et al (1997) Site alpha is crucial for two routes of IFN gamma-induced MHC class I transactivation: the ISRE-mediated route and a novel pathway involving CIITA. Immunity 6(5):601–611

    CAS  PubMed  Google Scholar 

  61. Bukur J et al (2010) Identification of E2F1 as an important transcription factor for the regulation of tapasin expression. J Biol Chem 285(40):30419–30426

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Zheng P et al (1998) Proto-oncogene PML controls genes devoted to MHC class I antigen presentation. Nature 396(6709):373–376

    CAS  PubMed  Google Scholar 

  63. Geiser AG et al (1993) Transforming growth factor beta 1 (TGF-beta 1) controls expression of major histocompatibility genes in the postnatal mouse: aberrant histocompatibility antigen expression in the pathogenesis of the TGF-beta 1 null mouse phenotype. Proc Natl Acad Sci USA 90(21):9944–9948

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Baldeon ME et al (1997) Interferon-gamma independently activates the MHC class I antigen processing pathway and diminishes glucose responsiveness in pancreatic beta-cell lines. Diabetes 46(5):770–778

    CAS  PubMed  Google Scholar 

  65. Israel A et al (1989) TNF stimulates expression of mouse MHC class I genes by inducing an NF kappa B-like enhancer binding activity which displaces constitutive factors. EMBO J 8(12):3793–3800

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Komov L et al (2018) Cell surface MHC class I expression is limited by the availability of peptide-receptive “empty” molecules rather than by the supply of peptide ligands. Proteomics 18(12):e1700248

    PubMed  Google Scholar 

  67. Parker BS, Rautela J, Hertzog PJ (2016) Antitumour actions of interferons: implications for cancer therapy. Nat Rev Cancer 16(3):131–144

    PubMed  Google Scholar 

  68. Schroder K et al (2004) Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol 75(2):163–189

    CAS  PubMed  Google Scholar 

  69. Ting JP, Baldwin AS (1993) Regulation of MHC gene expression. Curr Opin Immunol 5(1):8–16

    CAS  PubMed  Google Scholar 

  70. Hertzog PJ, Williams BR (2013) Fine tuning type I interferon responses. Cytokine Growth Factor Rev 24(3):217–225

    CAS  PubMed  Google Scholar 

  71. Zhou F (2009) Molecular mechanisms of IFN-gamma to up-regulate MHC class I antigen processing and presentation. Int Rev Immunol 28(3–4):239–260

    CAS  PubMed  Google Scholar 

  72. Strehl B et al (2005) Interferon-gamma, the functional plasticity of the ubiquitin-proteasome system, and MHC class I antigen processing. Immunol Rev 207:19–30

    CAS  PubMed  Google Scholar 

  73. Meissner TB et al (2010) NLR family member NLRC5 is a transcriptional regulator of MHC class I genes. Proc Natl Acad Sci USA 107(31):13794–13799

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Kulski JK et al (2001) Genomic and phylogenetic analysis of the human CD1 and HLA class I multicopy genes. J Mol Evol 53(6):642–650

    CAS  PubMed  Google Scholar 

  75. Gobin SJ, van den Elsen PJ (1999) The regulation of HLA class I expression: is HLA-G the odd one out? Semin Cancer Biol 9(1):55–59

    CAS  PubMed  Google Scholar 

  76. Gobin SJ et al (2002) HLA-G transactivation by cAMP-response element-binding protein (CREB) An alternative transactivation pathway to the conserved major histocompatibility complex (MHC) class I regulatory routes. J Biol Chem 277(42):39525–39531

    CAS  PubMed  Google Scholar 

  77. Flajollet S et al (2009) RREB-1 is a transcriptional repressor of HLA-G. J Immunol 183(11):6948–6959

    CAS  PubMed  Google Scholar 

  78. Eichmuller SB et al (2017) Immune modulatory microRNAs involved in tumor attack and tumor immune escape. J Natl Cancer Inst 109(10):1–14

    Google Scholar 

  79. Seliger B (2008) Different regulation of MHC class I antigen processing components in human tumors. J Immunotoxicol 5(4):361–367

    CAS  PubMed  Google Scholar 

  80. Kulkarni S et al (2011) Differential microRNA regulation of HLA-C expression and its association with HIV control. Nature 472(7344):495–498

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Tan Z et al (2007) Allele-specific targeting of microRNAs to HLA-G and risk of asthma. Am J Hum Genet 81(4):829–834

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Zhu XM et al (2010) Overexpression of miR-152 leads to reduced expression of human leukocyte antigen-G and increased natural killer cell mediated cytolysis in JEG-3 cells. Am J Obstet Gynecol 202(6):592

    PubMed  Google Scholar 

  83. Jasinski-Bergner S et al (2016) Identification of novel microRNAs regulating HLA-G expression and investigating their clinical relevance in renal cell carcinoma. Oncotarget 7(18):26866–26878

    PubMed  PubMed Central  Google Scholar 

  84. Friedrich M et al (2017) The role of the miR-148/-152 family in physiology and disease. Eur J Immunol 47(12):2026–2038

    CAS  PubMed  Google Scholar 

  85. Manaster I et al (2012) MiRNA-mediated control of HLA-G expression and function. PLoS ONE 7(3):e33395

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Jasinski-Bergner S et al (2015) Clinical relevance of miR-mediated HLA-G regulation and the associated immune cell infiltration in renal cell carcinoma. Oncoimmunology 4(6):e1008805

    PubMed  PubMed Central  Google Scholar 

  87. Wang Y et al (2017) MicroRNA-152 regulates immune response via targeting B7-H1 in gastric carcinoma. Oncotarget 8(17):28125–28134

    PubMed  PubMed Central  Google Scholar 

  88. Gao F et al (2013) miR-9 modulates the expression of interferon-regulated genes and MHC class I molecules in human nasopharyngeal carcinoma cells. Biochem Biophys Res Commun 431(3):610–616

    CAS  PubMed  Google Scholar 

  89. Bartoszewski R et al (2011) The unfolded protein response (UPR)-activated transcription factor X-box-binding protein 1 (XBP1) induces microRNA-346 expression that targets the human antigen peptide transporter 1 (TAP1) mRNA and governs immune regulatory genes. J Biol Chem 286(48):41862–41870

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Albanese M et al (2016) Epstein–Barr virus microRNAs reduce immune surveillance by virus-specific CD8 + T cells. Proc Natl Acad Sci USA 113(42):E6467–E6475

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Kim S et al (2011) Human cytomegalovirus microRNA miR-US4-1 inhibits CD8(+) T cell responses by targeting the aminopeptidase ERAP1. Nat Immunol 12(10):984–991

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Huang L et al (2018) The RNA-binding protein MEX3B mediates resistance to cancer immunotherapy by downregulating HLA-A expression. Clin Cancer Res 24(14):3366–3376

    CAS  PubMed  Google Scholar 

  93. Cano F et al (2012) The RNA-binding E3 ubiquitin ligase MEX-3C links ubiquitination with MHC-I mRNA degradation. EMBO J 31(17):3596–3606

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Reches A et al (2016) HNRNPR regulates the expression of classical and nonclassical MHC class I proteins. J Immunol 196(12):4967–4976

    CAS  PubMed  Google Scholar 

  95. Shankaran V et al (2001) IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 410(6832):1107–1111

    CAS  PubMed  Google Scholar 

  96. Kaplan DH et al (1998) Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc Natl Acad Sci USA 95(13):7556–7561

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Ivashkiv LB, Donlin LT (2014) Regulation of type I interferon responses. Nat Rev Immunol 14(1):36–49

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Greenlund AC et al (1994) Ligand-induced IFN gamma receptor tyrosine phosphorylation couples the receptor to its signal transduction system (p91). EMBO J 13(7):1591–1600

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Decker T et al (1991) Cytoplasmic activation of GAF, an IFN-gamma-regulated DNA-binding factor. EMBO J 10(4):927–932

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Chatterjee-Kishore M et al (2000) How Stat1 mediates constitutive gene expression: a complex of unphosphorylated Stat1 and IRF1 supports transcription of the LMP2 gene. EMBO J 19(15):4111–4122

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Rettino A, Clarke NM (2013) Genome-wide identification of IRF1 binding sites reveals extensive occupancy at cell death associated genes. J Carcinog Mutagen (Spec Iss Apoptosis). https://doi.org/10.4172/2157-2518.S6-009

    Article  Google Scholar 

  102. Starr R et al (1997) A family of cytokine-inducible inhibitors of signalling. Nature 387(6636):917–921

    CAS  PubMed  Google Scholar 

  103. Castro F et al (2018) Interferon-Gamma at the crossroads of tumor immune surveillance or evasion. Front Immunol 9:847

    PubMed  PubMed Central  Google Scholar 

  104. Seliger B et al (1997) IFN-gamma-mediated coordinated transcriptional regulation of the human TAP-1 and LMP-2 genes in human renal cell carcinoma. Clin Cancer Res 3(4):573–578

    CAS  PubMed  Google Scholar 

  105. Seliger B, Ruiz-Cabello F, Garrido F (2008) IFN inducibility of major histocompatibility antigens in tumors. Adv Cancer Res 101:249–276

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Respa A et al (2011) Association of IFN-gamma signal transduction defects with impaired HLA class I antigen processing in melanoma cell lines. Clin Cancer Res 17(9):2668–2678

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Aqbi HF et al (2018) IFN-gamma orchestrates tumor elimination, tumor dormancy, tumor escape, and progression. J Leukoc Biol. https://doi.org/10.1002/JLB.5MIR0917-351R

    Article  PubMed  Google Scholar 

  108. Schaefer L et al (2017) Proteoglycan neofunctions: regulation of inflammation and autophagy in cancer biology. FEBS J 284(1):10–26

    CAS  PubMed  Google Scholar 

  109. Recktenwald CV et al (2008) Altered detoxification status and increased resistance to oxidative stress by K-ras transformation. Cancer Res 68(24):10086–10093

    CAS  PubMed  Google Scholar 

  110. Recktenwald CV et al (2012) HER-2/neu-mediated down-regulation of biglycan associated with altered growth properties. J Biol Chem 287(29):24320–24329

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Subbarayan K et al (2018) Biglycan-mediated upregulation of MHC class I expression in HER-2/neu-transformed cells. Oncoimmunology 7(4):e1373233

    PubMed  PubMed Central  Google Scholar 

  112. Subbarayan K, Seliger B (2018) Tumor-dependent effects of proteoglycans and various glycosaminoglycan synthesizing enzymes and sulfotransferases on patients’ outcome. Curr Cancer Drug Targets 19(3):210–221

    Google Scholar 

  113. Yan L, DeMars LC (2012) Dietary supplementation with methylseleninic acid, but not selenomethionine, reduces spontaneous metastasis of Lewis lung carcinoma in mice. Int J Cancer 131(6):1260–1266

    CAS  PubMed  Google Scholar 

  114. Chen YC et al (2013) Dietary selenium supplementation modifies breast tumor growth and metastasis. Int J Cancer 133(9):2054–2064

    CAS  PubMed  Google Scholar 

  115. Labunskyy VM, Hatfield DL, Gladyshev VN (2014) Selenoproteins: molecular pathways and physiological roles. Physiol Rev 94(3):739–777

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Lennicke C et al (2017) Modulation of MHC class I surface expression in B16F10 melanoma cells by methylseleninic acid. Oncoimmunology 6(6):e1259049

    PubMed  Google Scholar 

  117. Kukita K et al (2015) Cancer-associated oxidase ERO1-alpha regulates the expression of MHC class I molecule via oxidative folding. J Immunol 194(10):4988–4996

    CAS  PubMed  Google Scholar 

  118. Paz-Ares L et al (2018) Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med 379(21):2040–2051

    CAS  PubMed  Google Scholar 

  119. Koshkin VS, Grivas P (2018) Emerging role of immunotherapy in advanced urothelial carcinoma. Curr Oncol Rep 20(6):48

    PubMed  Google Scholar 

  120. Boussiotis VA (2016) Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med 375(18):1767–1778

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Wolchok JD et al (2017) Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 377(14):1345–1356

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Sharma P et al (2017) Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168(4):707–723

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Barkal AA et al (2018) Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat Immunol 19(1):76–84

    CAS  PubMed  Google Scholar 

  124. Haworth KB et al (2015) Going back to class I: MHC and immunotherapies for childhood cancer. Pediatr Blood Cancer 62(4):571–576

    CAS  PubMed  Google Scholar 

  125. Chowell D et al (2018) Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 359(6375):582–587

    CAS  PubMed  Google Scholar 

  126. Goodman AM et al (2017) Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther 16(11):2598–2608

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Le DT et al (2017) Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357(6349):409–413

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Yeon Yeon S et al (2019) Immune checkpoint blockade resistance-related B2M hotspot mutations in microsatellite-unstable colorectal carcinoma. Pathol Res Pract 215(1):209–214

    CAS  PubMed  Google Scholar 

  129. Sade-Feldman M et al (2017) Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat Commun 8(1):1136

    PubMed  PubMed Central  Google Scholar 

  130. Anagnostou V et al (2017) Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer. Cancer Discov 7(3):264–276

    CAS  PubMed  Google Scholar 

  131. Ayers M et al (2017) IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 127(8):2930–2940

    PubMed  PubMed Central  Google Scholar 

  132. Zaretsky JM et al (2016) Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 375(9):819–829

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Budczies J et al (2017) Mutation patterns in genes encoding interferon signaling and antigen presentation: a pan-cancer survey with implications for the use of immune checkpoint inhibitors. Genes Chromosomes Cancer 56(8):651–659

    CAS  PubMed  Google Scholar 

  134. Ye Z et al (2018) Prevalent homozygous deletions of type I interferon and defensin genes in human cancers associate with immunotherapy resistance. Clin Cancer Res 24(14):3299–3308

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Paulson KG et al (2018) Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat Commun 9(1):3868

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Marijt KA, Doorduijn EM, van Hall T (2018) TEIPP antigens for T-cell based immunotherapy of immune-edited HLA class I(low) cancers. Mol Immunol. https://doi.org/10.1016/j.molimm.2018.03.029

    Article  PubMed  Google Scholar 

  137. Koyama S et al (2016) Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 7:10501

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Chojnacki S et al (2017) Programmatic access to bioinformatics tools from EMBL-EBI update: 2017. Nucleic Acids Res 45(W1):W550–W553

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Belmont PJ et al (2012) Regulation of microRNA expression in the heart by the ATF6 branch of the ER stress response. J Mol Cell Cardiol 52(5):1176–1182

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Colangelo T et al (2016) Proteomic screening identifies calreticulin as a miR-27a direct target repressing MHC class I cell surface exposure in colorectal cancer. Cell Death Dis 7:e2120

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Hisaoka M, Matsuyama A, Nakamoto M (2012) Aberrant calreticulin expression is involved in the dedifferentiation of dedifferentiated liposarcoma. Am J Pathol 180(5):2076–2083

    CAS  PubMed  Google Scholar 

  142. Zhao S et al (2015) MicroRNA-148a inhibits the proliferation and promotes the paclitaxel-induced apoptosis of ovarian cancer cells by targeting PDIA3. Mol Med Rep 12(3):3923–3929

    CAS  PubMed  Google Scholar 

  143. Mari L et al (2018) microRNA 125a regulates MHC-I expression on esophageal adenocarcinoma cells, associated with suppression of antitumor immune response and poor outcomes of patients. Gastroenterology 155(3):784–798

    CAS  PubMed  Google Scholar 

  144. Liu Y et al (2009) Altered expression profiles of microRNAs in a stable hepatitis B virus-expressing cell line. Chin Med J (Engl) 122(1):10–14

    CAS  Google Scholar 

  145. Kulkarni S et al (2017) Posttranscriptional regulation of HLA-A protein expression by alternative polyadenylation signals involving the RNA-binding protein syncrip. J Immunol 199(11):3892–3899

    CAS  PubMed  Google Scholar 

  146. Nachmani D et al (2014) MicroRNA editing facilitates immune elimination of HCMV infected cells. PLoS Pathog 10(2):e1003963

    PubMed  PubMed Central  Google Scholar 

  147. Kulkarni S et al (2013) Genetic interplay between HLA-C and MIR148A in HIV control and Crohn disease. Proc Natl Acad Sci USA 110(51):20705–20710

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Yin P et al (2015) MiR-451 suppresses cell proliferation and metastasis in A549 lung cancer cells. Mol Biotechnol 57(1):1–11

    CAS  PubMed  Google Scholar 

  149. Knox B et al (2018) A functional SNP in the 3′-UTR of TAP2 gene interacts with microRNA hsa-miR-1270 to suppress the gene expression. Environ Mol Mutagen 59(2):134–143

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Maria Heise for excellent secretarial help in preparing the manuscript.

Funding

This work was funded by the German Research Foundation (DFG; SE 581/22-1 and RTG, 1591/2-B4), the German Israeli Foundation for Scientific Research and Development (GIF; I-37-414.11-2016), and the Mildred Scheel Foundation.

Author information

Authors and Affiliations

Authors

Contributions

Barbara Seliger planned the manuscript. All the authors contributed in writing parts of the manuscript. Michael Friedrich, Simon Jasinski-Bergner, Barbara Seliger discussed the contents, while Michael Friedrich created the figures.

Corresponding author

Correspondence to Barbara Seliger.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This paper is a Focussed Research Review based on a presentation given at the Fourteenth Workshop & Symposium “Tumor Immunology meets Oncology” (TIMO XIV), held in Halle (Saale), Germany, 24th–26th May 2018. It is part of a series of Focussed Research Reviews and meeting report in Cancer Immunology, Immunotherapy.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Friedrich, M., Jasinski-Bergner, S., Lazaridou, MF. et al. Tumor-induced escape mechanisms and their association with resistance to checkpoint inhibitor therapy. Cancer Immunol Immunother 68, 1689–1700 (2019). https://doi.org/10.1007/s00262-019-02373-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-019-02373-1

Keywords

Navigation