1,629
Views
100
CrossRef citations to date
0
Altmetric
Reviews

Cellular and molecular mechanisms in cancer immune escape: a comprehensive review

&

References

  • Penn I. Cancer is a complication of severe immunosuppression. Surg. Gynecol. Obstet. 162, 603–610 (1986).
  • Fraumeni JF, Joseph F, Copeland G et al. Spectrum of Cancer Risk Among US Solid Organ Transplant Recipients. JAMA 306(17), 1891–1901 (2011).
  • Leeuwen MTV, Webster AC, Mccredie MRE et al. Effect of reduced immunosuppression after kidney transplant failure on risk of cancer: population based retrospective cohort study. BMJ 340, 570 (2010).
  • Shiels MS, Pfeiffer RM, Gail MH et al. Cancer burden in the HIV infected population in the United States. J. Natl Cancer Inst. 103(9), 753–62 (2011).
  • Rolston KVI, Bodey GP. Infections in Patients with Cancer. Holland-Frei Cancer Medicine (6th Edition). Bast RC, Kufe DW, Pollock RE, Weichselbaum RR, Holland JF, Frei E ( Eds). BC Decker, Hamilton, ON, USA (2000).
  • Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu. Rev. Immunol. 22, 329–360 (2004).
  • Burnet M. Cancer - a biological approach i. the processes of control. ii. the significance of somatic mutation. Br. Med. J. 1(5022), 779–786 (1957).
  • Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing Tumour development and shaping Tumour immunogenicity. Adv. Immunol. 90, 1–50 (2006).
  • Aris M, Barrio MM, Mordoh J. Lessons from Cancer Immunoediting in Cutaneous Melanoma. Clin. Dev. Immunol. 2012, 1–14 (2012).
  • Mocellin S, Nitti D. Therapeutics targeting tumor immune escape: towards the development of new generation anticancer vaccines. Med. Res. Rev. 28(3), 413–444 (2008).
  • Kontani K, Taguchi O, Narita T et al. Modulation of MUC1 mucin as an escape mechanism of breast cancer cells from autologous cytotoxic T-lymphocytes. Br. J. Cancer 84(9), 1258–1264 (2001).
  • Höchst B, Schildberg FA, Böttcher J et al. Liver sinusoidal endothelial cells contribute to CD8 T cell tolerance towards circulating carcinoembryonic antigen in mice. Hepatology 56(5), 1924–1933 (2012).
  • Bai XF, Liu J, Li O, Zheng P, Liu Y. Antigenic drift as a mechanism for tumour evasion of destruction by cytolytic T lymphocytes. J. Clin. Invest. 111(10), 1487–1496 (2003).
  • DuPage M, Mazumdar C, Schmidt LM et al. Expression of tumour-specific antigens underlies cancer immunoediting. Nature 482, 405–410 (2012).
  • Zhou G, Lu Z, McCadden JD, Levitsky HI, Marson AL. Reciprocal changes in tumour antigenicity and antigen-specific t-cell function during tumour progression. J Exp. Med. 200, 1581–1592 (2004).
  • Espana L, Fernandez Y, Rubio N, Torregrosa A, Blanco J, Sierra A. Overexpression of Bcl-xL in human breast cancer cells enhances organ-selective lymph node metastasis. Breast Cancer Res. Treat. 87, 33–44 (2004).
  • Jung YJ, Kim JY, Park JH. TGF-h1 inhibits Fas mediated apoptosis by regulating surface Fas and cFLIPL expression in human leukaemia/lymphoma cells. Int. J. Mol. Med. 13, 99–104 (2004).
  • Santiago B, Galindo M, Palao G, Pablos JL. Intracellular regulation of Fas-induced apoptosis in human fibroblasts by extracellular factors and cycloheximide. J. Immunol. 172, 560–566 (2004).
  • Volkmann M, Schiff JH, Hajjar Y et al. Loss of CD95 expression is linked to most but not all p53 mutants in European hepatocellular carcinoma. J. Mol. Med. 79, 594–600 (2001).
  • Maeda T, Yamada Y, Moriuchi R et al. Fas gene mutation in the progression of adult T-cell leukemia. J. Exp. Med. 189, 1063–1071 (1999).
  • Midis GP, Shen Y, Owen-Schaub LB. Elevated soluble Fas (sFas) levels in nonhematopoietic human malignancy. Cancer Res. 56, 3870–3874 (1996).
  • Ugurel S, Rappl G, Tilgen W, Reinhold U. Increased soluble CD95 (sFas/CD95) serum level correlates with poor prognosis in melanoma patients. Clin. Cancer Res. 7, 1282–1286 (2001).
  • Soriano C, Mukaro V, Hodge G et al. Increased proteinase inhibitor-9 (PI-9) and reduced granzyme B in lung cancer: Mechanism for immune evasion? Lung Cancer 77, 38–45 (2012).
  • Pitti RM, Marsters SA, Lawrence DA et al. Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature 396, 699–703 (1998).
  • Eaves CJ. Cancer stem cells: Here, there, everywhere? Nature 456(7222), 581–582 (2008).
  • Cheng J, Li L, Liu Y et al. Interleukin-1α induces immunesuppression by mesenchymal stem cells promoting the growth of prostate cancer cells. Mol. Med. Rep. 6, 955–960 (2012).
  • Schatton T, Schütte U, Frank NY et al. Modulation of T cell activation by malignant melanoma initiating cells. Cancer Res. 70, 697–708 (2010).
  • Mitchem JB, Brennan DJ, Knolhoff BL et al. Targeting Tumour-infiltrating macrophages decreases Tumour-Initiating cells, relieves immunosuppression and improves chemotherapeutic responses. Cancer Res. 73(3), 1128–1141 (2013).
  • Noh KH, Kim BW, Song KH et al. Nanog signaling in cancer promotes stem-like phenotype and immune evasion. J. Clin. Invest. 122, 4077–4093 (2012).
  • Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 4, 891–899 (2004).
  • Cairns RA, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat. Rev. Cancer 11, 85–95 (2011).
  • Calcinotto A, Filipazzi P, Grioni M et al. Modulation of microenvironment acidity reverses anergy in human and murine Tumour-infiltrating T lymphocytes. Cancer Res. 72, 2746–2756 (2012).
  • Bellone M, Calcinotto A, Filipazzi P, De Milito A, Fais S, Rivoltini L. The acidity of the Tumour microenvironment is a mechanism of immune escape that can be overcome by proton pump inhibitors. Oncoimmunology 1, e22058 (2013).
  • Markus D, Jacek RW. Hypoxia inducible factor-1 in cancer immune suppression. Curr. Immunol. Rev. 6, 260–271 (2010).
  • Uyttenhove C, Pilotte L, Theate I et al. Evidence for a Tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat. Med. 9, 1269–1274 (2003).
  • Allarino F, Grohmann U, You S et al. The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T-cell receptor ζ-chain and induce a regulatory phenotype in naive T cells. J. Immunol. 176, 6752–6761 (2006).
  • Prendergast GC, Metz R, Muller AJ. Towards a Genetic Definition of Cancer-Associated Inflammation: role of the IDO Pathway. Am. J. Pathol. 176, 2082–2087 (2010).
  • Villablanca E, Godin-Ethier J, Hanafi LA, Piccirillo CA, Lapointe R. Indoleamine 2,3-Dioxygenase Expression in Human Cancers: Clinical and Immunologic Perspectives. Clin. Cancer Res. 17(22), 6985–6991 (2011).
  • Seton-Rogers S. Metabolism: Catabolic effects. Nat. Rev. Cancer 11, 757 (2011).
  • Herber DL, Cao W, Nefedova Y et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat. Med. 16, 880–886 (2010).
  • Villablanca EJ, Raccosta L, Zhou D et al. Tumour-mediated liver X receptor-α activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens anti tumour responses. Nat. Med. 16, 98–105 (2010).
  • Wang D, Dubois RN. Prostaglandins and cancer. Gut 55, 115–122 (2006).
  • Sharma S, Stolina M, Yang SC et al. Tumour cyclooxygenase 2-dependent suppression of dendritic cell function. Clin. Cancer Res. 9, 961–968 (2003).
  • Yao C, Sakata D, Esaki Y et al. Prostaglandin E2-EP4 signalling promotes immune inflammation through Th1 cell differentiation and Th17 cell expansion. Nat. Med. 15, 633–640 (2009).
  • Sharma S, Yang SH, Zhu L et al. Tumour cyclooxygenase-2/prostaglandin E2-dependent promotion of FOXP3 expression and CD4 CD25 T regulatory cell activities in lung cancer. Cancer Res. 65, 5211–5220 (2005).
  • Rodriguez PC, Hernandez CP, Quiceno D et al. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J. Exp. Med. 202, 931–939 (2005).
  • Moussalli MJ, Wu Y, Zuo X et al. Mechanistic Contribution of Ubiquitous 15-Lipoxygenase-1 Expression Loss in Cancer Cells to Terminal Cell Differentiation Evasion. Cancer Prev. Res. (Phila) 4, 1961–1972 (2011).
  • Hirata T, Yamamoto H, Taniguchi H et al. Characterization of the immune escape phenotype of human gastric cancers with and without high-frequency microsatellite instability. J. Pathol. 211(5), 516–523 (2007).
  • Sadun RE, Sachsman SM, Chen X et al. Immune Signatures of Murine and Human Cancers Reveal Unique Mechanisms of Tumour Escape and New Targets for Cancer Immunotherapy. Clin. Cancer Res. 13, 4016–4025 (2007).
  • Bidwell BN, Slaney CY, Withana NP et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 18, 1224–1231 (2012).
  • Mouillot G, Marcou C, Rousseau P et al. HLA-G gene activation in Tumour cells involves cis-acting epigenetic changes. Int. J. Cancer. 113, 928–936 (2005).
  • Serrano A, Castro-Vega I, Redondo M. Role of gene methylation in antitumor immune response: implication for tumor progression. Cancers 3, 1672–1690 (2011).
  • Murtas D, Maric D, Giorgi VD et al. IRF-1 responsiveness to IFN-ã predicts different cancer immune phenotypes. Br. J. Cancer 109, 76–82 (2013).
  • Stern-Ginossar N, Gur C, Biton M et al. Human microRNAs regulate stress-induced immune responses mediated by the receptor NKG2D. Nat. Immunol. 9, 1065–1073 (2008).
  • Heinz M, Lemke P, Anagnostopoulos I et al. Nuclear factor kappa B-dependent gene expression profiling of Hodgkin’s disease Tumour cells, pathogenetic significance, and link to constitutive signal transducer and activator of transcription 5a activity. J. Exp. Med. 196, 605–617 (2002).
  • Chen R, Alvero AB, Silasi DA, Mor G. Inflammation, cancer and chemoresistance: taking advantage of the toll-like receptor signalling pathway. Am. J. Reprod. Immunol. 57, 93–107 (2007).
  • Bentires-Alj M, Barbu V, Fillet M et al. NF-kappa B transcription factor induces drug resistance through MDR1 expression in cancer cells. Oncogene 22, 90–97 (2003).
  • Schoof N, Bonin FV, Trümper L, Kube D. HSP90 is essential for Jak-STAT signaling in classical Hodgkin lymphoma cells. Cell Commun. Sig. 7, 17 (2009).
  • Yu H, Kortylewski M, Pardoll D. Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Mol. Immunol. 44, 2850–2859 (2007).
  • Kortylewski M, Kujawski M, Wang T et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumour immunity. Nat. Med. 11, 1314–1321 (2005).
  • Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 441, 424–430 (2006).
  • Yuan TL, Cantley LC. PI3K pathway alterations in cancer: variations on a theme. Oncogene 27, 5497–5510 (2008).
  • Noh KH, Kang TH, Kim JH et al. Activation of AKT as a mechanism for tumour immune evasion. Mol. Ther. 17, 439–447 (2009).
  • Sumimoto H, Imabayashi F, Iwata T, Kawakami Y. The BRAF–MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 203, 1651–1656 (2006).
  • He W, Liu Q, Wang L, Chen W, Li N, Cao X. TLR4 signaling promotes immune escape of human lung cancer cells by inducing immunosuppressive cytokines and apoptosis resistance. Mol. Immunol. 44, 2850–2859 (2007).
  • Szczepanski MJ, Czystowska M, Szajnik M et al. Triggering of Toll like receptor 4 expressed on human head and neck squamous cell carcinoma promotes Tumour development and protects the Tumour from immune attack. Cancer Res. 69, 3105–3113 (2009).
  • Sun Z, Luo Q, Ye D, Chen W, Chen F. Role of toll-like receptor 4 on the immune escape of human oral squamous cell carcinoma and resistance of cisplatin-induced apoptosis. Mol. Cancer. 11, 33–44 (2012).
  • Calderwood SK, Murshid A, Gong J. Heat shock proteins: conditional mediators of inflammation in tumour immunity. Front. Immunol. 3, 1–10 (2012).
  • Dempsey NC, Leoni F, Ireland HE, Hoyle C, Williams JHH. Differential heat shock protein localization in chronic lymphocytic leukemia. J. Leukoc. Biol. 87, 467–476 (2010).
  • Jameel A, Skilton RA, Campbell TA, Chander SK, Coombes RC, Luqmani YA. Clinical and biological significance of Hsp90a in human breast cancer. Int. J. Cancer 50, 409–415 (1992).
  • Yano M, Naito Z, Tanaka S, Asano G. Expression and roles of heat shock proteins in human breast cancer. Jpn J. Cancer Res. 87, 908–915 (1996).
  • Ciocca DR, Calderwood SK. Heat shock proteins in cancer: diagnostic, prognostic, predictive, and treatment implications. Cell Stress Chaperones 10, 86–103 (2005).
  • Beere HM, Wolf BB, Cain K et al. Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome. Nat. Cell Biol. 2, 469–475 (2000).
  • Saleh A, Srinivasula SM, Balkir L, Robbins PD, Alnemri ES. Negative regulation of the Apaf-1 apoptosome by Hsp70. Nat. Cell Biol. 2, 476–483 (2000).
  • Vanden Berghe T, Kalai M, van Loo G, Declercq W, Vandenabeele P. Disruption of Hsp90 function reverts Tumour necrosis factor-induced necrosis to apoptosis. J. Biol. Chem. 278, 5622–5629 (2003).
  • Chen G, Cao P, Goeddel DV. TNF-induced recruitment and activation of the IKK complex require cdc37 and Hsp90. Mol. Cell. 9, 401–410 (2002).
  • Nathan DE, Vos MH, Lindquist S. In vivo functions of the Saccharomyces cerevisiae Hsp90 chaperone. Proc. Natl Acad. Sci. USA 94, 12949–12956 (1997).
  • Pratt WB. The Hsp90-based chaperone system: involvement in signal transduction from a variety of hormone and growth factor receptors. Proc. Soc. Exp. Biol. Med. 217, 420–431 (1998).
  • Blagosklonny MV, Toretsky J, Bohen S, Neckers L. Mutant conformation of p53 translated in vitro or in vivo requires functional Hsp90. Proc. Natl Acad. Sci. USA 93, 8379–8383 (1996).
  • Banerjee S, Lin CFL, Skinner KA et al. Heat shock protein 27 differentiates tolerogenic macrophages that may support human breast cancer progression. Cancer Res. 71, 318–327 (2011).
  • Lizee G, Radvanyi LG, Overwijk WW, Hwu P. Improving antitumour immune responses by circumventing immunoregulatory cells and mechanisms. Clin. Cancer Res. 12, 4794–4803 (2006).
  • Yang Ma, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J. Cancer 4, 36–44 (2013).
  • Esche C, Lokshin A, Shurin GV et al. Tumour’s other immune targets: dendritic cells. J. Leukoc. Biol. 66, 336–344 (1999).
  • Kiertscher SM, Luo J, Dubinett SM, Roth MD. Tumours promote altered maturation and early apoptosis of monocyte-derived dendritic cells. J. Immunol. 164, 1269–1276 (2000).
  • Peguet-Navarro J, Sportouch M, Popa I, Berthier O, Schmitt D, Portoukalian J. Gangliosides from human melanoma Tumours impair dendritic cell differentiation from monocytes and induce their apoptosis. J. Immunol. 170, 3488–3494 (2003).
  • Yang T, Witham TF, Villa L et al. Glioma-associated hyaluronan induces apoptosis in dendritic cells via inducible nitric oxide synthase: implications for the use of dendritic cells for therapy of gliomas. Cancer Res. 62, 2583–2591 (2002).
  • Lu J, Aggarwal R, Kanji S et al. Human Ovarian Tumor Cells Escape γδ T Cell Recognition Partly by Down Regulating Surface Expression of MICA and Limiting Cell Cycle Related Molecules. PLoS ONE 6(9), e23348 (2011).
  • Kim R, Emi M, Tanabe K, Uchida Y, Toge T. The role of Fas ligand and transforming growth factor β in Tumour progression: molecular mechanisms of immune privilege via Fas-mediated apoptosis and potential targets for cancer therapy. Cancer 100, 2281–2291 (2004).
  • Young MRI, Wright MA, Coogan M, Young ME, Bagash J. Tumour-derived cytokines induce bone marrow suppressor cells that mediate immunosuppression through transforming growth factor β. Cancer Immunol. Immunother. 35, 14–18 (1992).
  • Gabrilovich DI, Nadaf S, Corak J, Berzofsky JA, Carbone DP. Dendritic cells in antitumour immune responses. II. Dendritic cells grown from bone marrow precursors, but not mature DC from Tumour-bearing mice, are effective antigen carriers in the therapy of established Tumours. Cell Immunol. 170, 111–119 (1996).
  • Chaux P, Moutet M, Faivre J, Martin F, Martin M. Inflammatory cells infiltrating human colorectal carcinomas express HLA class II but not B7-1 and B7-2 costimulatory molecules of the T-cell activation. Lab. Invest. 74, 975–983 (1996).
  • Tourkova IL, Shurin GV, Wei S, Shurin MR. Small rho GTPases mediate tumour-induced inhibition of endocytic activity of dendritic cells. J. Immunol. 178, 7787–7793 (2007).
  • Tourkova IL, Shurin GV, Chatta GS et al. Restoration by IL-15 of MHC class I antigen-processing machinery in human dendritic cells inhibited by Tumour-derived gangliosides. J. Immunol. 175, 3045–3052 (2005).
  • Maleno I, Cabrera CM, Cabrera T et al. Distribution of HLA class I altered phenotypes in colorectal carcinomas: high frequency of HLA haplotype loss associated with loss of heterozygosity in chromosome region 6p21. Immunogenetics 56, 244–253 (2004).
  • Menon AG, Morreau H, Tollenaar RA et al. Down-regulation of HLA-A expression correlates with a better prognosis in colorectal cancer patients. Lab. Invest. 82, 1725–1733 (2002).
  • Browning M, Petronzelli F, Bicknell D et al. Mechanisms of loss of HLA class I expression on colorectal Tumour cells. Tissue Antigens 47, 364–371 (1996).
  • Hanagiri T, Shigematsu Y, Kuroda K et al. Prognostic implications of human leukocyte antigen class I expression in patients who underwent surgical resection for non-small cell lung cancer. J. Surg. Res. 181, e57–e63 (2013).
  • Garrido F, Ruiz-Cabello F, Cabrera T et al. Implications for immunosurveillance of altered HLA class I phenotypes in human tumours. Immunol. Today 18(2), 89–95 (1997).
  • Cabrera T, Pedrajas G, Cozar JM et al. HLA class I expression in bladder carcinomas. Tissue Antigens 62(4), 324–327 (2003).
  • Challa-Malladi M, Lieu YK, Califano O et al. Combined Genetic Inactivation of β2-Microglobulin and CD58 Reveals Frequent Escape from Immune Recognition in Diffuse Large B Cell. Cancer Cell. 20, 728–740 (2011).
  • Steidl C, Shah SP, Woolcock BW et al. MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers. Nature 471, 377–81 (2011).
  • Oldford SA, Robb JD, Codner D, Gadag V, Watson PH, Drover S. Tumor cell expression of HLA-DM associates with a Th1 profile and predicts improved survival in breast carcinoma patients. Int. Immunol. 18, 1591–602 (2006).
  • Luijn MMV, Chamuleau MED, Ossenkoppele GJ, Loosdrecht AAV, Ham SMV. Tumor immune escape in acute myeloid leukemia: Class II -associated invariant chain peptide expression as result of deficient antigen presentation. Oncoimmunol 1(2), 211–213 (2012).
  • Frolich D, Blabetafeld D, Reiter K et al. The anti-CD74 humanized monoclonal antibody, milatuzumab, which targets the invariant chain of MHC II complexes, alters B-cell proliferation, migration, and adhesion molecule expression. Arthritis Res. Ther. 14, R54 (2012).
  • Theobald M, Biggs J, Hernandez J et al. Tolerance to p53 by A2.1-restricted cytotoxic T lymphocytes. J. Exp. Med. 185, 833–841 (1997).
  • Zang X, Allison JP. The B7 family and cancer therapy: costimulation and coinhibition. Clin. Cancer Res. 13, 5271–5279 (2007).
  • Zang X, Lok P, Kim J, Murphy K, Waitz R, Allison JP. B7x: a widely expressed B7 family member that inhibits T-cell activation. Proc. Natl Acad. Sci. USA 100, 10388–10392 (2003).
  • Ebelt K, Babaryka G, Frankenberger B et al. Prostate cancer lesions are surrounded by FOXP3+, PD-1+ and B7-H1+ lymphocyte clusters. Eur. J. Cancer 45, 1664–1672 (2009).
  • Freeman GJ, Long AJ, Iwai Y et al. Engagement of the PD-1immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 192, 1027–1034 (2000).
  • Richards S, Mai XM, Dong C. B7S1, a novel B7 family member that negatively regulates T-cell activation. Immunity 18, 863–873 (2003).
  • Jeon H, Ohaegbulam KC, Abadi YM, Zang X. B7x and myeloid-derived suppressor cells in the tumor microenvironment: a tale of two cities. OncoImmunology 2(7), e24744 (2013).
  • Strand S, Hofmann WJ, Hug H et al. Lymphocyte apoptosis induced by CD95 (APO-1/Fas) ligand-expressing Tumour cells: A mechanism of immune evasion? Nat. Med. 2, 1361–1366 (1996).
  • Kim JW, Tsukishiro T, Johnson JT, Whiteside TL. Expression of pro- and antiapoptotic proteins in circulating CD8+ T-cells of patients with squamous cell carcinoma of the head and neck. Clin. Cancer Res. 10, 5101–5110 (2004).
  • Reichert TE, Strauss L, Wagner EM, Gooding W, Whiteside TL. Signaling abnormalities, apoptosis, and reduced proliferation of circulating and Tumour-infiltrating lymphocytes in patients with oral carcinoma. Clin. Cancer Res. 8, 3137–3145 (2002).
  • Kim W-J, Tsukishiro T, Johnson JT, Whiteside TL. Expression of pro- and ant-apoptotic proteins in circulating CD8+ T-cells of patients with squamous cell carcinoma of the head and neck. Clin. Cancer Res. 10, 5101–5110 (2004).
  • Dong H, Strome SE, Salomao DR et al. Tumour-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat. Med. 8, 793–800 (2002).
  • Nakashima M, Sonoda K, Watanabe T. Inhibition of cell growth and induction of apoptotic cell death by the human Tumour-associated antigen RCAS1. Nat. Med. 5, 938–942 (1999).
  • Lopez CB, Rao TD, Feiner H, Shapiro R, Marks JR, Frey AB. Repression of IL-2 mRNA translation in primary human breast carcinoma Tumour-infiltrating lymphocytes. Cell Immunol. 190, 141–155 (1998).
  • Olkhanud PB, Damdinsuren B, Bodogai M et al. Tumour-Evoked Regulatory B-cells Promote Breast Cancer Metastasis by Converting Resting CD4+ T-cells to T-Regulatory Cells. Cancer Res. 71, 3505–3515 (2011).
  • Bodogai M, Chang CL, Wejksza K, Lai J, Merino M, Wersto RP et al. Anti-CD20 antibody promotes cancer escape via enrichment of tumour-evoked regulatory b-cells expressing low levels of CD20 and CD137L. Cancer Res 73, 2127(2013).
  • Costello RT, Gastaut JA, Olive D. Tumour escape from immune surveillance. Arch. Immunol. Ther. Exp. 47, 83–88 (1999).
  • Mamessier E, Sylvain A, Thibult ML et al. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumour immunity. J. Clin. Invest. 121, 3609–3622 (2011).
  • Fauriat C, Just-Landi S, Mallet F et al. Deficient expression of NCR in NK cells from acute myeloid leukemia: evolution during leukemia treatment and impact of leukemia cells in NCR dull phenotype induction. Blood 109(1), 323–330 (2007).
  • Maki G, Hayes GM, Naji A et al. NK resistance of Tumour cells from multiple myeloma and chronic lymphocytic leukemia patients: implication of HLA-G. Leukemia 22(5), 998–1006 (2008).
  • Groh V, Wu J, Yee C, Spies T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 419, 734–738 (2002).
  • Nakajima H, Zhao R, Lund TC et al. The BCR/ABL transgene causes abnormal NK cell differentiation and can be found in circulating NK cells of advanced phase chronic myelogenous leukemia patients. J. Immunol. 168, 643–650 (2002).
  • Wilson EB, El-Jawhari JJ, Neilson AA et al. Human tumour immune evasion via TGF-β blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity. PLoS ONE 6(9), e22842 (2011).
  • Hao N, Lü M, Fan Y et al. Macrophages in Tumor Microenvironments and the Progression of Tumors. Clin. Develop. Immunol. 1–11 (2012).
  • Quatromoni JG, Eruslanov E. Tumor-associated macrophages: function, phenotype, and link to prognosis in human lung cancer. Am. J. Transl Res. 4(4), 376–389 (2012).
  • Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 12, 253–268 (2012).
  • Kuang DM. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 206, 1327–1337 (2009).
  • Tazzyman S, Niaz H, Murdoch C. Neutrophil-mediated tumour angiogenesis: subversion of immune responses to promote tumour growth. Semin. Cancer Biol. 23(3), 149–158 (2013).
  • Klink M, Jastrzembska K, Nowak M et al. Ovarian cancer cells modulate human blood neutrophils response to activation in vitro. Scand. J. Immunol. 68(3), 328–336 (2008).
  • Cho H, Hur HW, Kim SW et al. Pre-treatment neutrophil to lymphocyte ratio is elevated in epithelial ovarian cancer and predicts survival after treatment. Cancer Immunol. Immunother. 58(1), 15–23 (2009).
  • Hix LM, Shi YH, Brutkiewicz RR et al. CD1d-Expressing Breast Cancer Cells Modulate NKT Cell-Mediated Antitumor Immunity in a Murine Model of Breast Cancer Metastasis. PLoS ONE 6(6), e20702 (2011).
  • Terabe M, Swann J, Ambrosino E et al. A nonclassical non-Valpha14Jalpha18 CD1d-restricted (type II) NKT-cell is sufficient for down-regulation of tumour immunosurveillance. J. Exp. Med. 202, 1627–1633 (2005).
  • Stewart TJ, Smyth MJ, Fernando GJ, Frazer IH, Leggatt GR. Inhibition of early Tumour growth requires J alpha 18-positive (natural killer T) cells. Cancer Res. 63, 3058–3060 (2003).
  • Lu J, Aggarwal R, Kanji S et al. Human Ovarian Tumor Cells Escape γδ T Cell Recognition Partly by Down Regulating Surface Expression of MICA and Limiting Cell Cycle Related Molecules. PLoS ONE 6(9), e23348 (2011).
  • Rodriguez PC, Quiceno DG, Zabaleta J et al. Arginase I production in the Tumour microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 64, 5839–5849 (2004).
  • Nagaraj S, Gabrilovich DI. Myeloid-derived suppressor cells in human cancer. Cancer J. 16, 348–353 (2010).
  • Rodríguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol. Rev. 222, 180–191 (2008).
  • Rodriguez PC, Zea AH, DeSalvo J et al. L-arginine consumption by macrophages modulates the expression of CD3 zeta chain in T lymphocytes. J. Immunol. 171, 1232–1239 (2003).
  • Rodriguez PC, Quiceno DG, Ochoa AC. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood 109, 1568–1573 (2007).
  • Bingisser RM, Tilbrook PA, Holt PG, Kees UR. Macrophage-derived nitric oxide regulates T-cell activation via reversible disruption of the Jak3/STAT5 signalling pathway. J. Immunol. 160, 5729–5734 (1998).
  • Harari O, Liao JK. Inhibition of MHC II gene transcription by nitric oxide and antioxidants. Curr. Pharm. Des. 10, 893–898 (2004).
  • Rivoltini L, Carrabba M, Huber V et al. Immunity to cancer: attack and escape in T lymphocyte-Tumour cell interaction. Immunol. Rev. 188, 97–113 (2002).
  • Nagaraj S, Gupta K, Pisarev V et al. Altered recognition of antigen is a mechanism of CD8+ T-cell tolerance in cancer. Nat. Med. 13, 828–835 (2007).
  • Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J. Immunol. 182, 4499–4506 (2009).
  • Sauer H, Wartenberg M, Hescheler J. Reactive oxygen species as intracellular messengers during cell growth and differentiation. Cell. Physiol. Biochem. 11, 173–186 (2001).
  • Yang L, Huang J, Ren X et al. Abrogation of TGF beta signalling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 13, 23–35 (2008).
  • Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 70, 68–77 (2010).
  • Hanson EM, Clements VK, Sinha P, Ilkovitch D, Ostrand-Rosenberg S. Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T-cells. J. Immunol. 183, 937–944 (2009).
  • Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand-Rosenberg S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts Tumour immunity toward a type 2 response. J. Immunol. 179, 977–983 (2007).
  • Yu J, Du W, Yan F et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J. Immunol. 190(7), 3783–3797 (2013).
  • Li H, Han Y, Guo Q, Zhang M, Cao X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta-1. J. Immunol. 182, 240–249 (2009).
  • Yang L, DeBusk LM, Fukuda K et al. Expansion of myeloid immune suppressor Gr+CD11b+ cells in Tumour-bearing host directly promotes Tumour angiogenesis. Cancer Cell. 6, 409–4021 (2004).
  • Ko JS, Bukowski RM, Fincke JH. Myeloid-derived suppressor cells: a novel therapeutic target. Curr. Oncol. Rep. 11, 87–93 (2009).
  • Chantrain CF, Shimada H, Jodele S et al. Stromal matrix metalloproteinase-9 regulates the vascular architecture in neuroblastoma by promoting pericyte recruitment. Cancer Res. 64, 1675–1686 (2004).
  • Sakaguchi S. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T-cells in immunological tolerance to self and non-self. Nat. Immunol. 6, 345–352 (2005).
  • Woo EY, Chu CS, Goletz TJ et al. Regulatory CD4(+)CD25(+) T-cells in Tumours from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Res. 61, 4766–4772 (2001).
  • Liyanage UK, Moore TT, Joo HG et al. Prevalence of regulatory T-cells is increased in peripheral blood and Tumour microenvironment of patients with pancreas or breast adenocarcinoma. J. Immunol. 169, 2756–2761 (2002).
  • Yan M, Jene N, Byrne D et al. Recruitment of regulatory T-cells is correlated with hypoxia-induced CXCR4 expression, and is associated with poor prognosis in basal-like breast cancers. Breast Cancer Res. 13, R47 (2011).
  • Facciabene A, Peng X, Hagemann IS et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature 475, 226–230 (2011).
  • Seo N, Hayakawa S, Takigawa M, Tokura Y. IL-10 expressed at early tumour sites induces subsequent generation of CD4(+) T-regulatory cells and systemic collapse of antitumour immunity. Immunology 103, 449–457 (2001).
  • Chen W, Jin W, Hardegen N et al. Conversion of peripheral CD4+CD25- naive T-cells to CD4+CD25+ regulatory T-cells by TGF-beta induction of transcription factor Foxp3. J. Exp. Med. 198,1875–1886 (2003).
  • Sato E, Olson SH, Ahn J et al. Intraepithelial CD8+ Tumour-infiltrating lymphocytes and a high CD8+/regulatory T-cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl Acad. Sci. USA 102, 18538–18543 (2005).
  • Vignali DA, Collison LW, Workman CJ. How regulatory T-cells work. Nat. Rev. Immunol. 8, 523–532 (2008).
  • Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J. Immunol. 174, 1783–1786 (2005).
  • Ren X, Ye F, Jiang Z, Chu Y, Xiong S, Wang Y. Involvement of cellular death in TRAIL/DR5-dependent suppression induced by CD4(+)CD25(+) regulatory T-cells. Cell Death Differ. 14, 2076–2084 (2007).
  • Garín MI, Chu CC, Golshayan D, Cernuda-Morollón E, Wait R, Lechler RI. Galectin-1: a key effector of regulation mediated by CD4+CD25+ T-cells. Blood 109, 2058–2065 (2007).
  • Cao X, Cai SF, Fehniger TA et al. Granzyme B and perforin are important for regulatory T-cell-mediated suppression of Tumour clearance. Immunity 27, 635–646 (2007).
  • Kim R, Emi M, Tanabe K, Uchida Y, Toge T. The role of Fas ligand and transforming growth factor β in Tumour progression: molecular mechanisms of immune privilege via Fas-mediated apoptosis and potential targets for cancer therapy. Cancer 100, 2281–2291 (2004).
  • Critchley-Thorne RJ, Simons DL, Yan N et al. Impaired interferon signaling is a common immune defect in human cancer. Proc. Natl Acad. Sci. USA 106(22), 9010–9015 (2009).
  • Rangel-Corona R, Corona-Ortega T, Soto-Cruz I et al. Evidence that cervical cancer cells secrete IL-2, which becomes an autocrine growth factor. Cytokine 50, 273–277 (2010).
  • Ramsay AG, Clear AJ, Fatah R et al. Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood 120(7), 1412–142 (2012).
  • T-Wu TC. The Role of Vascular Cell Adhesion Molecule-1 in Tumour Immune Evasion. Cancer Res. 67, 6003–6006 (2007).
  • Lippitz BE. Cytokine patterns in patients with cancer: a systematic review. Lancet Oncol. 14, e218–e228 (2013).
  • Wang L, Yi T, Kortylewski M, Pardoll DM, Zeng D, Yu H. IL-17 can promote tumour growth through an IL-6–Stat3 signalling pathway. J. Exp. Med. 206, 1457–1464 (2009).
  • Langowski JL, Zhang X, Wu L et al. IL-23 promotes tumour incidence and growth. Nature 442, 461–465 (2006).
  • Taylor DD, Gerçel-Taylor C. Tumour-derived exosomes and their role in cancer-associated T-cell signalling defects. Br. J. Cancer. 92, 305–311 (2005).
  • Taylor DD, Gerçel-Taylor C, Lyons KS, Stanson J, Whiteside TL. T-cell apoptosis and suppression of T-cell receptor/CD3-ζ by Fas ligand-containing membrane vesicles shed from ovarian tumors. Clin. Cancer Res. 9, 5113–5119 (2003).
  • Clayton A, Mitchell JP, Court J, Linnane S, Mason MD, Tabi Z. Human tumor derived exosomes down-modulate NKG2D expression. J. Immunol. 180, 7249–7258 (2008).
  • Klibi J, Niki T, Riedel A et al. Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood 113(9), 1957–1966 (2009).
  • Ohshima K, Inoue K, Fujiwara A et al. Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS ONE 5(10), e13247 (2010).
  • Huber V, Fais S, Iero M et al. Human colorectal cancer cells induce T-cell death through release of proapoptotic microvesicles: role in immune escape. Gastroenterology 128, 1796–1804 (2005).
  • Kollet O, Petit I, Kahn J et al. Human CD34+CXCR4−sorted cells harbour intracellular CXCR4, which can be functionally expressed and provide NOD/SCID repopulation. Blood 100, 2778–2786 (2002).
  • Kryczek I, Wei S, Keller E, Liu R, Zou W. Stroma-derived factor (SDF-1/CXCL12) and human Tumour pathogenesis. Am. J. Physiol. Cell. Physiol. 292(3), C987–C995 (2007).
  • Koshiba T, Hosotani R, Miyamoto Y et al. Expression of stromal cell-derived factor 1 and CXCR4 ligand receptor system in pancreatic cancer: a possible role for Tumour progression. Clin. Cancer Res. 6, 3530–3535 (2000).
  • Shields JD, Kourtis IC, Tomei AT, Roberts JM, Swartz MA. Induction of lymphoid like stroma and immune escape by Tumours that express the chemokine CCL21. Science 328(5979), 749–752 (2010).
  • Allard B, Turcotte M, Stagg J. CD73-generated adenosine: orchestrating the tumour-stroma interplay to promote cancer growth. J. Biomed. Biotech. 2012, 1–8 (2012).
  • Placke T, Oergel M, Schaller M et al. Platelet-derived MHC Class I confers a pseudo- normal phenotype to cancer cells that subverts the anti-Tumour reactivity of natural killer immune cells. Cancer Res. 72, 440–448 (2011).
  • Kopp HG, Placke T, Salih HR. Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumour reactivity. Cancer Res. 69, 7775–7783 (2009).
  • Hofmeister V, Schrama D, Becker JC. Anti-cancer therapies targeting the Tumour stroma. Cancer Immunol. Immunother. 57(1), 1–17 (2008).
  • Noman MZ, Buart S, Van PJ et al. The cooperative induction of hypoxia-inducible factor-1 alpha and STAT3 during hypoxia induced an impairment of Tumour susceptibility to CTL-mediated cell lysis. J. Immunol. 182, 3510–3521 (2009).
  • Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells. J. Immunol. 185(4), 2273–2284 (2010).
  • Mocellin S, Benna C, Pilati P. Coinhibitory molecules in cancer biology and therapy. Cytokine Growth Factor Rev. 24, 147–161 (2013).
  • Tarhini A, Lo E, Minor DR. Releasing the brake on the immune system: ipilimumab in melanoma and other tumors. Cancer Biother. Radiopharm. 25(6), 601–613 (2010).
  • Dangaj D, Lanitis E, Zhao A et al. Novel recombinant human B7-H4 antibodies overcome tumoral immune escape to potentiate T-cell anti-Tumour responses. Cancer Res. 73(15), 4820–4829 (2013).
  • Pastor F, Kolonias D, McNamara JO, Gilboa E. Targeting 4–1BB costimulation to disseminated tumor lesions with bi-specific oligonucleotide aptamers. Mol. Ther. 19(10), 1878–1886 (2011).
  • Barbara S, Ramos AC, Liu E et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor–modified T cells in lymphoma patients. J. Clin. Invest. 121(5), 1822–1826 (2011).
  • Melero I, Hirschhorn-Cymerman D, Morales-Kastresana A, Sanmamed MF, Wolchok JD. Agonist antibodies to TNFR molecules that costimulate T and NK cells. Clin. Cancer Res. 19(5), 1044–1053 (2013).
  • Cohen AD, Schaer DA, Liu C et al. Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS ONE 5, e10436 (2010).
  • Pedroza-Gonzalez A, Verhoef C, Ijzermans JN et al. Activated tumor-infiltrating CD4+ regulatory T cells restrain antitumor immunity in patients with primary or metastatic liver cancer. Hepatology 57(1), 183–194 (2013).
  • Ruter J, Barnett BG, Kryczek I et al. Altering regulatory T-cell function in cancer immunotherapy: a novel means to boost the efficacy of cancer vaccines. Front. Biosci. 14, 1761–1770 (2009).
  • Galustian C, Meyer B, Labarthe MC et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol. Immunother. 58, 1033–1045 (2009).
  • Finke JH, Rini B, Ireland J et al. Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clin. Cancer Res.14, 6674–6682 (2008).
  • Sun Q, Liu Q, Zheng Y, Cao X. Rapamycin suppresses TLR4-triggered IL-6 and PGE2 production of colon cancer cells by inhibiting TLR4 expression and NF-κB activation. Mol. Immunol. 45, 2929–2936 (2008).
  • Tkach M, Coria L, Rosemblit C et al. Targeting Stat3 induces senescence in tumor cells and elicits prophylactic and therapeutic immune responses against breast cancer growth mediated by NK cells and CD4+ T cells. J. Immunol. 189(3), 1162–1172 (2012).
  • Zhang L, Alizadeh D, Van Handel M, Kortylewski M, Yu H, Badie B. Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice. Glia 57(13), 1458–1467 (2009).
  • Kitamura H, Torigoe T, Asanuma H, Honma I, Sato N, Tsukamoto T. Down-regulation of hla class i antigens in prostate cancer tissues and up-regulation by histone deacetylase inhibition. J. Urol. 178(2), 692–696 (2007).
  • Khan ANH, Gregori CJ, Tomasi TB. Histone deacetylase inhibitors induce TAP, LMP, Tapasin genes and MHC class I antigen presentation by melanoma cells. Cancer Immunol. Immnuother. 57, 647–654 (2008).
  • Serrano A, Castro-Vega I, Redondo M. Role of gene methylation in antitumor immune response: implication for tumor progression. Cancers 3, 1672–1690 (2011).
  • Higano CS, Schellhammer PF, Small EJ et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer 115, 3670–3679 (2009).
  • Kantoff PW, Higano CS, Shore ND et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 363, 411–422 (2010).
  • Zheng X, Koropatnick J, Chen D et al. Silencing IDO in dendritic cells: a novel approach to enhance cancer immunotherapy in a murine breast cancer model. Int. J. Cancer. 132(4), 967–77 (2013).
  • Garg NK, Dwivedi P, Prabha P, Tyagi RK. RNA pulsed dendritic cells: An approach for cancer immunotherapy. Vaccine 31(8), 1141–1156 (2013).
  • Soliman HH, Antonia S, Sullivan D, Vanahanian N, Link C. Overcoming Tumour antigen anergy in human malignancies using the novel indoleamine 2,3-dioxygenase (IDO) enzyme inhibitor, 1-methyl-D-tryptophan (1MT). J. Clin. Oncol. 27, 15s (2009).
  • Lob S, Konigsrainer A, Schafer R, Rammensee HG, Opelz G, Terness P. Levo- but not dextro-1-methyl tryptophan abrogates the IDO activity of human dendritic cells. Blood 111, 2152–2154 (2008).
  • Frölich D, Blaβfeld D, Reiter K et al. The anti-CD74 humanized monoclonal antibody, milatuzumab, which targets the invariant chain of MHC II complexes, alters B-cell proliferation, migration, and adhesion molecule expression. Arth. Res. Ther. 14, R54 (2012).
  • Pacey S, Wilson RH, Walton M et al. A Phase I study of the Heat Shock Protein 90 inhibitor alvespimycin (17-DMAG) given intravenously to patients with advanced solid tumours. Clin. Cancer Res. 17(6), 1561–1570 (2011).
  • Rao A, Kawabe M, Storkus W. Integration of HSP90 inhibitors in combinational immunotherapy targeting receptor tyrosine kinases (RTKs) in cancer. J. Immunol. 184, 131.45 (2010).
  • Trepel J, Mollapour M, Giaccone G, Neckers L. Targeting the dynamic HSP90 complex in cancer. Nat. Rev. Cancer 10(8), 537–549 (2010).
  • Sangiolo D.. Cytokine induced killer cells as promising immunotherapy for solid tumours. J. Cancer 2, 363–368 (2011).
  • Teague RM, Sather BD, Sacks JA et al. Interleukin-15 rescues tolerant CD8+ T cells for use in adoptive immunotherapy of established tumors. Nat. Med. 12, 335–341 (2006).
  • Caserta S, Alessi P, Basso V, Mondino A. IL-7 is superior to IL-2 for ex vivo expansion of tumour-specific CD4 (+) T cells. Eur. J. Immunol. 40, 470–79 (2010).
  • Zaanen VHC, Lokhorst HM, Aarden LA, Rensink HJ, Warnaar SO, Van-Der Lelie J, Van-Oers MH. Chimaeric anti-interleukin 6 monoclonal antibodies in the treatment of advanced multiple myeloma: A phase I dose-escalating study. Br. J. Haematol. 102, 783–790 (1998).
  • Gorelik L, Flavell RA. Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat. Med. 7, 1118–1122 (2001).
  • Fakhrai H, Mantil JC, Liu L et al. Phase I clinical trial of a TGF-beta antisense-modified tumor cell vaccine in patients with advanced glioma. Cancer Gene Ther. 13, 1052–1060 (2006).
  • Nemunaitis J, Dillman RO, Schwarzenberger PO et al. Phase II study of belagenpumatucel-L, a transforming growth factor beta-2antisense gene-modified allogeneic tumor cell vaccine in non-small-cell lung cancer. J Clin. Oncol. 24, 4721–4730 (2006).
  • Vega EA, Graner MW, Sampson JH. Combating immunosuppression in glioma. Fut. Oncol. 4(3), 433–442 (2008).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.