73
Views
5
CrossRef citations to date
0
Altmetric
Hypothesis

“High Treg” Inflammations Promote (Most) Non-Hematologic Cancers While “Low Treg” Inflammations Promote Lymphoid Cancers

Pages 209-221 | Published online: 21 May 2020

References

  • Elkoshi Z. The binary classification of chronic diseases. J Inflamm Res. 2019;12:319–333. doi:10.2147/JIR.S22727931908517
  • Sugimoto MA, Sousa LP, Pinho V, Perretti M, Teixeira MM. Resolution of inflammation: what controls its onset? Front Immunol. 2016;7:160. doi:10.3389/fimmu.2016.0016027199985
  • Todoric J, Antonucci L, Karin M. Targeting inflammation in cancer prevention and therapy. Cancer Prev Res (Phila). 2016;9(12):895–905. doi:10.1158/1940-6207.CAPR-16-020927913448
  • Ritter B, Greten FR. Modulating inflammation for cancer therapy. J Exp Med. 2019;216(6):1234–1243. doi:10.1084/jem.2018173931023715
  • Son WC, Gopinath C. Early occurrence of spontaneous tumors in CD-1 mice and sprague—dawley rats. Toxicol Pathol. 2004;32(4):371–374. doi:10.1080/0192623049044087115307208
  • Deng G. Tumor-infiltrating regulatory T cells: origins and features. Am J Clin Exp Immunol. 2018;7(5):81–87.30498624
  • Buckner JH. Mechanisms of impaired regulation by CD4+CD25+FOXP3+regulatory T cells in human autoimmune diseases. Nat Rev Immunol. 2010;10(12):849–859. doi:10.1038/nri288921107346
  • Shull MM, Ormsby I, Kier AB, et al. Targeted disruption of the mouse transforming growth factor-β 1 gene results in multifocal inflammatory disease. Nature. 1992;359:693–699. doi:10.1038/359693a01436033
  • Dang H, Geiser AG, Letterio JJ, et al. SLE-like autoantibodies and Sjogren’s syndrome-like lymphoproliferation in TGF-β knockout mice. J Immunol. 1995;155:3205–3212.7673733
  • Tada Y, Togashi Y, Kotani D, et al. Targeting VEGFR2 with Ramucirumab strongly impacts effector/activated regulatory T cells and CD8+ T cells in the tumor microenvironment. J Immunother Cancer. 2018;6(1):106. doi:10.1186/s40425-018-0403-130314524
  • Wu F, Weigel KJ, Zhou H, Wang XJ. Paradoxical roles of TGF-β signaling in suppressing and promoting squamous cell carcinoma. Acta Biochim Biophys Sin (Shanghai). 2018;50(1):98–105. doi:10.1093/abbs/gmx12729206939
  • Ohue Y, Nishikawa H. Regulatory T (Treg) cells in cancer: can Treg cells be a new therapeutic target? Cancer Sci. 2019;110(7):2080–2089. doi:10.1111/cas.1406931102428
  • Fujimura T, Kambayashi Y, Aiba S. Crosstalk between regulatory T cells (Tregs) and myeloid derived suppressor cells (MDSCs) during melanoma growth. Oncoimmunology. 2012;1(8):1433–1434. doi:10.4161/onci.2117623243619
  • Zhang C, Wang S, Yang C, Rong R. The crosstalk between myeloid derived suppressor cells and immune cells: to establish immune tolerance in transplantation. J Immunol Res. 2016;2016:4986797. doi:10.1155/2016/498679727868073
  • Singh S, Mehta N, Lilan J, Budhthoki MB, Chao F, Yong L. Initiative action of tumor-associated macrophage during tumor metastasis. Biochim Open. 2017;4:8–18. doi:10.1016/j.biopen.2016.11.00229450136
  • Kornete M, Piccirillo CA. Functional crosstalk between dendritic cells and Foxp3(+) regulatory T cells in the maintenance of immune tolerance. Front Immunol. 2012;3:165. doi:10.3389/fimmu.2012.0016522737152
  • Wylie W, Macri M, Mintern M, Waithman W. Dendritic cells and cancer: from biology to therapeutic intervention. Cancers (Basel). 2019;11(4):521. doi:10.3390/cancers11040521
  • Li XC, Wang MY, Yang M, et al. A mutational signature associated with alcohol consumption and prognostically significantly mutated driver genes in esophageal squamous cell carcinoma. Ann Oncol. 2018;29(4):938–944. doi:10.1093/annonc/mdy01129351612
  • Li X, Xu W, Kang W, et al. Genomic analysis of liver cancer unveils novel driver genes and distinct prognostic features. Theranostics. 2018;8(6):1740–1751. doi:10.7150/thno.2201029556353
  • Osna NA, Donohue TM Jr, Kharbanda KK. Alcoholic liver disease: pathogenesis and current management. Alcohol Res. 2017;38(2):147–161.28988570
  • Holt AP, Salmon M, Buckley CD, Adams DH. Immune interactions in hepatic fibrosis. Clin Liver Dis. 2008;12(4):861–82, x. doi:10.1016/j.cld.2008.07.00218984471
  • Fabregat I, Moreno-Càceres J, Sánchez A, et al. TGF-β signalling and liver disease. FEBS J. 2016;283(12):2219–2232. doi:10.1111/febs.1366526807763
  • Prystupa A, Kiciński P, Sak J, et al. Proinflammatory cytokines (IL-1α, IL-6) and hepatocyte growth factor in patients with alcoholic liver cirrhosis. Gastroenterol Res Pract. 2015;2015:532615. doi:10.1155/2015/53261526448742
  • Zhang G. The role of transforming growth factor β in T helper 17 differentiation. Immunology. 2018;155(1):24–35. doi:10.1111/imm.1293829682722
  • Sagiv JY, Michaeli J, Assi S, et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 2015;10(4):562–573. doi:10.1016/j.celrep.2014.12.03925620698
  • Lin TH, Shao YY, Chan SY, Huang CY, Hsu CH, Cheng AL. High serum transforming growth factor-β1 levels predict outcome in hepatocellular carcinoma patients treated with sorafenib. Clin Cancer Res. 2015;21(16):3678–3684. doi:10.1158/1078-043225977342
  • Albillos A, Lario M, Álvarez-Mon M. Cirrhosis-associated immune dysfunction: distinctive features and clinical relevance. J Hepatol. 2014;61(6):1385–1396. doi:10.1016/j.jhep.2014.08.01025135860
  • Yu LX, Ling Y, Wang HY. Role of nonresolving inflammation in hepatocellular carcinoma development and progression. NPJ Precis Oncol. 2018;2(1):6. doi:10.1038/s41698-018-0048-z29872724
  • Mou H, Wu S, Zhao G, Wang J. Changes of Th17/Treg ratio in the transition of chronic hepatitis B to liver cirrhosis and correlations with liver function and inflammation. Exp Ther Med. 2019;17(4):2963–2968. doi:10.3892/etm.2019.729930936966
  • Zhao HQ, Li WM, Lu ZQ, Yao YM. Roles of tregs in development of hepatocellular carcinoma: a meta-analysis. World J Gastroenterol. 2014;20(24):7971–7978. doi:10.3748/wjg.v20.i24.797124976734
  • Van Herck MA, Weyler J, Kwanten WJ, et al. The differential roles of T cells in non-alcoholic fatty liver disease and obesity. Front Immunol. 2019;10:82. doi:10.3389/fimmu.2019.0008230787925
  • Sakurai T, Kudo M. Molecular link between liver fibrosis and hepatocellular carcinoma. Liver Cancer. 2013;2(3–4):365–366. doi:10.1159/00034385124400223
  • Flecken T, Schmidt N, Hild S, et al. Immunodominance and functional alterations of tumor-associated antigen-specific CD8+ T-cell responses in hepatocellular carcinoma. Hepatology. 2014;59(4):1415–1426. doi:10.1002/hep.2673124002931
  • Kim YK, Lee BC, Ham BJ, et al. Increased transforming growth factor-beta1 in alcohol dependence. J Korean Med Sci. 2009;24(5):941–944. doi:10.3346/jkms.2009.24.5.94119794996
  • Bagnardi V, Rota M, Botteri E, et al. Alcohol consumption and site-specific cancer risk: a comprehensive dose-response meta-analysis. Br J Cancer. 2015;112(3):580–593. doi:10.1038/bjc.2014.57925422909
  • Alexandrov LB, Ju YS, Haase K, et al. Mutational signatures associated with tobacco smoking in human cancer. Science. 2016;354(6312):618–622. doi:10.1126/science.aag02927811275
  • Chiba T, Chihara J, Furue M. Role of the Arylhydrocarbon Receptor (AhR) in the pathology of asthma and COPD. J Allergy (Cairo). 2012;2012:372384. doi:10.1155/2012/37238422500183
  • Lugade AA, Bogner PN, Thatcher TH, Sime PJ, Phipps RP, Thanavala Y. Cigarette smoke exposure exacerbates lung inflammation and compromises immunity to bacterial infection. J Immunol. 2014;192(11):5226–5235. doi:10.4049/jimmunol.130258424752444
  • Ostadkarampour M, Müller M, Öckinger J, et al. Distinctive regulatory T cells and altered cytokine profile locally in the airways of young smokers with normal lung function. PLoS One. 2016;11(10):e0164751. doi:10.1371/journal.pone.016475127798682
  • Ward H. Oxford Handbook of Epidemiology for Clinicians. Oxford University Press;2012:289–290. ISBN 978-0-19-165478-7
  • Kalathil SG, Lugade AA, Pradhan V, et al. T-Regulatory cells and programmed death 11 T cells contribute to effector T-cell dysfunction in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2014;190(1):40–50. doi:10.1164/rccm.201312-2293OC24825462
  • Bhat TA, Panzica L, Kalathil SG, Thanavala Y. Immune dysfunction in patients with chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2015;12(Suppl 2):S169–S175. doi:10.1513/AnnalsATS.201503-126AW26595735
  • Wang J, Linxweiler M, Yang W, Chan TA, Morris LGT. Immunomodulatory and immunotherapeutic implications of tobacco smoking in squamous cell carcinomas and normal airway epithelium. Oncotarget. 2019;10(39):3835–3839. doi:10.18632/oncotarget.2698231231463
  • Prado-Garcia H, Romero-Garcia S, Aguilar-Cazares D, Meneses-Flores M, Lopez-Gonzalez JS. Tumor-induced CD8+ T-cell dysfunction in lung cancer patients. Clin Dev Immunol. 2012;2012:741741. doi:10.1155/2012/74174123118782
  • Wang JB, Huang X, Li FR. Impaired dendritic cell functions in lung cancer: a review of recent advances and future perspectives. Cancer Commun. 2019;39(1):43. doi:10.1186/s40880-019-0387-3
  • Gandini S, Botteri E, Iodice S, et al. Tobacco smoking and cancer: a meta‐analysis. Int J Cancer. 2008;122(1):155–164. doi:10.1002/ijc.2303317893872
  • Sergentanis TN, Kanavidis P, Michelakos T, Petridou ET. Cigarette smoking and risk of lymphoma in adults: a comprehensive meta-analysis on hodgkin and non-hodgkin disease. Eur J Cancer Prev. 2013;22(2):131–150. doi:10.1097/CEJ.0b013e328355ed0822759975
  • Tzankov A, Meier C, Hirschmann P, Went P, Pileri SA, Dirnhofer S. Correlation of high numbers of intratumoral FOXP3+ regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical hodgkin’s lymphoma. Haematologica. 2008;93(2):193–200. doi:10.3324/haematol.1170218223287
  • Alvaro T, Lejeune M, Salvadó MT, et al. Outcome in hodgkin’s lymphoma can be predicted from the presence of accompanying cytotoxic and regulatory T cells. Clin Cancer Res. 2005;11(4):1467–1473. doi:10.1158/1078-0432.CCR-04-186915746048
  • Engels EA. Infectious agents as causes of non-Hodgkin lymphoma. Cancer Epidemiol Biomarkers Prev. 2007;16(3):401–404. doi:10.1158/1055-9965.EPI-06-105617337646
  • Edwards LA, Nistala K, Mills DC, et al. Delineation of the innate and adaptive T-cell immune outcome in the human host in response to Campylobacter jejuni infection. PLoS One. 2010;5(11):e15398. doi:10.1371/journal.pone.001539821085698
  • Anderson LA, Atman AA, McShane CM, et al. Common infection-related conditions and risk of lymphoid malignancies in older individuals. Br J Cancer. 2014;110(11):2796–2803. doi:10.1038/bjc.2014.17324691420
  • Cunningham MW. Rheumatic fever, autoimmunity, and molecular mimicry: the streptococcal connection. Int Rev Immunol. 2014;33(4):314–329. doi:10.3109/08830185.2014.91741124892819
  • Peveling-Oberhag J, Arcaini L, Hansmann ML, Zeuzem S. Hepatitis C-associated B-cell non-hodgkin lymphomas. Epidemiology, molecular signature and clinical management. J Hepatol. 2013;59(1):169–177. doi:10.1016/j.jhep.2013.03.01823542089
  • Rivera-Correa JRA. In Malaria: Immune Response to Infection and Vaccination (Eds Rodriguez, A. & Mota, M.). Switzerland: Springer; 2016.
  • Rivera-Correa J, Guthmiller JJ, Vijay R, et al. Plasmodium DNA-mediated TLR9 activation of T-bet+ B cells contributes to autoimmune anaemia during malaria. Nat Commun. 2017;8(1):1282. doi:10.1038/s41467-017-01476-629101363
  • Landgren O, Engels EA, Pfeiffer RM, et al. Autoimmunity and susceptibility to hodgkin lymphoma: a population-based case-control study in Scandinavia. J Natl Cancer Inst. 2006;98(18):1321–1330. doi:10.1093/jnci/djj36116985251
  • Li P, Zheng Y, Chen X. Drugs for autoimmune inflammatory diseases: from small molecule compounds to anti-TNF biologics. Front Pharmacol. 2017;8:460. doi:10.3389/fphar.2017.0046028785220
  • Bereshchenko O, Coppo M, Bruscoli S, et al. GILZ promotes production of peripherally induced Treg cells and mediates the crosstalk between glucocorticoids and TGF-b signaling. Cell Rep. 2014;7(2):464–475. doi:10.1016/j.celrep.2014.03.00424703841
  • Pufall MA. Glucocorticoids and cancer. Adv Exp Med Biol. 2015;872:315–333. doi:10.1007/978-1-4939-2895-8_1426216001
  • Tao R, de Zoeten EF, Ozkaynak E, et al. Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med. 2007;13:1299–1307. doi:10.1038/nm165217922010
  • Ge Z, Da Y, Xue Z, et al. Vorinostat, a histone deacetylase inhibitor, suppresses dendritic cell function and ameliorates experimental autoimmune encephalomyelitis. Exp Neurol. 2013;241:56–66. doi:10.1016/j.expneurol.2012.12.00623261766
  • Christensen DP, Gysemans C, Lundh M, et al. Lysine deacetylase inhibition prevents diabetes by chromatin-independent immunoregulation and β-cell protection. Proc Natl Acad Sci U S A. 2014;111(3):1055–1059. doi:10.1073/pnas.132085011124395784
  • Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J, Kaupper T, Roncarolo MG. Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients. J Immunol. 2006;177(12):8338–8347. doi:10.4049/jimmunol.177.12.833817142730
  • Eriksson P, Wallin P, Sjöwall C. Clinical experience of sirolimus regarding efficacy and safety in systemic lupus erythematosus. Front Pharmacol. 2019;10:82. doi:10.3389/fphar.2019.0008230787878
  • Teachey DT, Greiner R, Seif A, et al. Treatment with sirolimus results in complete responses in patients with autoimmune lymphoproliferative syndrome. Br J Haematol. 2009;145(1):101–106. doi:10.1111/j.1365-2141.2009.07595.x19208097
  • Jasinski S, Weinblatt ME, Glasser CL. Sirolimus as an effective agent in the treatment of immune thrombocytopenia (ITP) and Evans Syndrome (ES): a single institution’s experience. J Pediatr Hematol Oncol. 2017;39(6):420–424. doi:10.1097/MPH.000000000000081828267088
  • Armand P, Gannamaneni S, Kim HT, et al. Improved survival in lymphoma patients receiving sirolimus for graft-versus-host disease prophylaxis after allogeneic hematopoietic stem-cell transplantation with reduced-intensity conditioning. J Clin Oncol. 2008;26(35):5767–5774. doi:10.1200/JCO.2008.17.727919001324
  • Carlé A, Pedersen IB, Knudsen N, et al. Moderate alcohol consumption may protect against overt autoimmune hypothyroidism: a population-based case-control study. Eur J Endocrinol. 2012;167(4):483–490. doi:10.1530/EJE-12-035622802427
  • Papageorgis P, Stylianopoulos T. Role of TGFβ in regulation of the tumor microenvironment and drug delivery (Review). Int J Oncol. 2015;46(3):933–943. doi:10.3892/ijo.2015.281625573346
  • Newcom SR, Kadin ME, Ansari AA. Production of transforming growth factor-beta activity by Ki-1 positive lymphoma cells and analysis of its role in the regulation of Ki-1 positive lymphoma growth. Am J Pathol. 1988;131(3):569–577.2898211
  • Kadin ME, Cavaille-Coll MW, Gertz R, Massagué J, Cheifetz S, George D. Loss of receptors for transforming growth factor beta in human T-cell malignancies. Proc Natl Acad Sci U S A. 2019;10(39):3835–3839. doi:10.1073/pnas.91.13.6002
  • Lockmer S, Wahlin BE, Smedby KE, Kimby E. Chemotherapy-free initial treatment of advanced indolent lymphoma has durable effect with low toxicity: results from two nordic lymphoma group trials with more than 10 years of follow-up. J Clin Oncol. 2018;36(33):3315–3323. doi:10.1200/JCO.18.00262
  • Muñoz-Antonia T, Muro-Cacho C, Sharma S, Cantor A, Bepler G. Expression of TGFbeta type-II receptor in association with markers of proliferation and apoptosis in premalignant lung lesions. Cancer. 2007;110(7):1527–1531. doi:10.1002/cncr.2293717676583
  • Anderson M, Muro-Cacho C, Cordero J, Livingston S, Muñoz-Antonia T. Transforming growth factor beta receptors in verrucous and squamous cell carcinoma. Arch Otolaryngol Head Neck Surg. 1999;125(8):849–854. doi:10.1001/archotol.125.8.84910448730
  • Gobbi H, Dupont WD, Simpson JF, et al. Transforming growth factor-beta and breast cancer risk in women with mammary epithelial hyperplasia. J Natl Cancer Inst. 1999;91(24):2096–2101. doi:10.1093/jnci/91.24.209610601380
  • Shang B, Liu Y, Jiang SJ, Liu Y. Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Sci Rep. 2015;5:15179. doi:10.1038/srep1517926462617
  • Vacchelli E, Semeraro M, Enot DP, et al. Negative prognostic impact of regulatory T cell infiltration in surgically resected esophageal cancer post-radiochemotherapy. Oncotarget. 2015;6(25):20840–20850. doi:10.18632/oncotarget.442826369701
  • Wrzesinski SH, Wan YY, Flavell RA. Transforming growth factor-beta and the immune response: implications for anticancer therapy. Clin Cancer Res. 2007;13(18 Pt 1):5262–5270. doi:10.1158/1078-0432.CCR-07-115717875754
  • Nakagaki T, Tamura M, Kobashi K, et al. Profiling cancer-related gene mutations in oral squamous cell carcinoma from Japanese patients by targeted amplicon sequencing. Oncotarget. 2017;8(35):59113–59122. doi:10.18632/oncotarget.1926228938622
  • Tokunaga H, Lee DH, Kim IY, Wheeler TM, Lerner SP. Decreased expression of transforming growth factor b receptor type I Is associated with poor prognosis in bladder transitional cell carcinoma patients. Clin Cancer Res. 1999;5(9):2520–2525.10499628
  • Winerdal ME, Marits P, Winerdal M, et al. FOXP3 and survival in urinary bladder cancer. BJU Int. 2011;108:1672–1678. doi:10.1111/j.1464-410X.2010.10020.x21244603
  • Bakkebø M, Huse K, Hilden VI, Smeland EB, Oksvold MP. TGF-b-induced growth inhibition in B-cell lymphoma correlates with Smad1/5 signalling and constitutively active p38 MAPK. BMC Immunol. 2010;11:57. 2010. doi:10.1186/1471-2172-11-57
  • Wang B, Dileepan T, Briscoe S, et al. Induction of TGF-β1 and TGF-β1–dependent predominant Th17 differentiation by group A streptococcal infection. Proc Natl Acad Sci U S A. 2010;107(13):5937–5942. doi:10.1073/pnas.090483110720231435
  • Costalonga M, Cleary PP, Fischer LA, et al. Intranasal bacteria induce Th1 but not Treg or Th2. Mucosal Immunol. 2009;2(1):85–95. doi:10.1038/mi.2008.6719079337
  • Linnebacher M, Maletzki C, Klier U, Klar E. Bacterial immunotherapy of gastrointestinal tumors. Langenbecks Arch Surg. 2012;397(4):557–568. doi:10.1007/s00423-011-0892-622189906