77
Views
3
CrossRef citations to date
0
Altmetric
Hypothesis

The Binary Classification of Protein Kinases

Pages 929-947 | Published online: 18 Mar 2021

References

  • Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298(5600):1912–1934. doi:10.1126/science.1075762
  • Ardito F, Giuliani M, Perrone D, Troiano G, Lo Muzio L. The crucial role of protein phosphorylation in cell signaling and its use as targeted therapy (review). Int J Mol Med. 2017;40(2):271–280. doi:10.3892/ijmm.2017.3036
  • Sacco F, Perfetto L, Castagnoli L, Cesareni G. The human phosphatase interactome: an intricate family portrait. FEBS Lett. 2012;586(17):2732–2739. doi:10.1016/j.febslet.2012.05.008
  • Ferguson FM, Gray NS. Kinase inhibitors: the road ahead. Nat Rev Drug Discov. 2018;17(5):353–377. doi:10.1038/nrd.2018.21
  • Szilveszter KP, Németh T, Mócsai A. Tyrosine kinases in autoimmune and inflammatory skin diseases. Front Immunol. 2019;10:1862. doi:10.3389/fimmu.2019.01862
  • Siasos G, Tousoulis D, Siasou Z, Stefanadis C, Papavassiliou AG. Shear stress, protein kinases and atherosclerosis. Curr Med Chem. 2007;14(14):1567–1572. doi:10.2174/092986707780831087
  • Kinase-disease association/cell signaling technology. Available from: https://www.cellsignal.com/contents/resources-reference-tables/kinase-disease-associations/science-tables-kinase-disease. Accessed February 26, 2021.
  • Elkoshi Z. The binary classification of chronic diseases. J Inflamm Res. 2019;12:319–333. doi:10.2147/JIR.S227279
  • Elkoshi Z. “High treg” inflammations promote (most) non-hematologic cancers while “low treg” inflammations promote lymphoid cancers. J Inflamm Res. 2020;13:209–221. doi:10.2147/JIR.S249384
  • 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.2014.579
  • 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-12-0356
  • Zhang X, Fang X, Gao Z, et al. Axitinib, a selective inhibitor of vascular endothelial growth factor receptor, exerts an anticancer effect in melanoma through promoting antitumor immunity. Anticancer Drugs. 2014;25(2):204–211. doi:10.1097/CAD.0000000000000033
  • Napoletano C, Ruscito I, Bellati F, et al. Bevacizumab-based chemotherapy triggers immunological effects in responding multi-treated recurrent ovarian cancer patients by favoring the recruitment of effector T cell subsets. J Clin Med. 2019;8(3):380. doi:10.3390/jcm8030380
  • Bergerot P, Lamb P, Wang E, Pal SK. Cabozantinib in combination with immunotherapy for advanced renal cell carcinoma and urothelial carcinoma: rationale and clinical evidence. Mol Cancer Ther. 2019;18(12):2185–2193. doi:10.1158/1535-7163.MCT-18-1399
  • Kwilas AR, Ardiani A, Donahue RN, Aftab DT, Hodge JW. Dual effects of a targeted small-molecule inhibitor (cabozantinib) on immune-mediated killing of tumor cells and immune tumor microenvironment permissiveness when combined with a cancer vaccine. J Transl Med. 2014;12(1):294. doi:10.1186/s12967-014-0294-y
  • Jie HB, Schuler PJ, Lee SC, et al. CTLA-4+ regulatory T cells increased in cetuximab-treated head and neck cancer patients suppress NK cell cytotoxicity and correlate with poor prognosis. Cancer Res. 2015;75(11):2200–2210. doi:10.1158/0008-5472.CAN-14-2788
  • Liu P, Zhao L, Kepp O, Kroemer G. Crizotinib - a tyrosine kinase inhibitor that stimulates immunogenic cell death. Oncoimmunology. 2019;8(7):1596652. doi:10.1080/2162402X.2019.1596652
  • Fei F, Yu Y, Schmitt A, et al. Dasatinib inhibits the proliferation and function of CD4+CD25+ regulatory T cells. Br J Haematol. 2009;144(2):195–205. doi:10.1111/j.1365-2141.2008.07433.x
  • Palakurthi S, Kuraguchi M, Zacharek SJ, et al. The combined effect of FGFR inhibition and PD-1 blockade promotes tumor-intrinsic induction of antitumor immunity. Cancer Immunol Res. 2019;7(9):1457–1471. doi:10.1158/2326-6066.CIR-18-0595
  • Ayeni D, Miller B, Kuhlmann A, et al. Tumor regression mediated by oncogene withdrawal or erlotinib stimulates infiltration of inflammatory immune cells in EGFR mutant lung tumors. J Immunother Cancer. 2019;7(1):172. doi:10.1186/s40425-019-0643-8
  • Wang S, Zhang Y, Wang Y, et al. Amphiregulin confers regulatory T cell suppressive function and tumor invasion via the EGFR/GSK-3β/Foxp3 axis. J Biol Chem. 2016;291(40):21085–21095. doi:10.1074/jbc.M116.717892
  • Podhorecka M, Goracy A, Szymczyk A, et al. Changes in T-cell subpopulations and cytokine network during early period of ibrutinib therapy in chronic lymphocytic leukemia patients: the significant decrease in T regulatory cells number. Oncotarget. 2017;8(21):34661–34669. doi:10.18632/oncotarget.16148
  • Larmonier N, Janikashvili N, LaCasse CJ, et al. Imatinib mesylate inhibits CD4+ CD25+ regulatory T cell activity and enhances active immunotherapy against BCR-ABL- tumors. J Immunol. 2008;181(10):6955–6963. doi:10.4049/jimmunol.181.10.6955
  • Kimura T, Kato Y, Ozawa Y, et al. Immunomodulatory activity of lenvatinib contributes to antitumor activity in the Hepa1-6 hepatocellular carcinoma model. Cancer Sci. 2018;109(12):3993–4002. doi:10.1111/cas.13806
  • Lu Z, Xu N, Zhou X, et al. Therapeutic immune monitoring of CD4+CD25+ T cells in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors. Oncol Lett. 2017;14(2):1363–1372. doi:10.3892/ol.2017.6294
  • Wang X, Shan S, Semba T, et al. Abstract 4492: humanized anti-EGFR antibody panitumumab inhibits tumor growth of inflammatory breast cancer by inducing antitumor immunity. Cancer Res. 2019;79(13 Supplement):4492. doi:10.1158/1538-7445.AM2019-4492
  • Verzoni E, De Cecco L, Dugo M, et al. Broad immunomodulating effect of first-line pazopanib in metastatic renal cell carcinoma patients. Ann Oncol. 2017:28. doi:10.1093/annonc/mdx371.043.
  • Busse A, Asemissen AM, Nonnenmacher A, et al. Immunomodulatory effects of sorafenib on peripheral immune effector cells in metastatic renal cell carcinoma. Eur J Cancer. 2011;47(5):690–696. doi:10.1016/j.ejca.2010.11.021
  • Stehle F, Schulz K, Fahldieck C, et al. Reduced immunosuppressive properties of axitinib in comparison with other tyrosine kinase inhibitors. J Biol Chem. 2013;288(23):16334–16347. doi:10.1074/jbc.M112.437962
  • 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. 2008;14(20):6674–6682. doi:10.1158/1078-0432.CCR-07-5212
  • Salas RN, Finke JH, Rini B. The intersection of sunitinib with the immunosuppressive microenvironment of renal cell carcinoma: implications for future therapeutics. Target Oncol. 2007;2(4):225–234. doi:10.1007/s11523-007-0064-3
  • Ko JS, Zea AH, Rini BI, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res. 2009;15(6):2148–2157. doi:10.1158/1078-0432.CCR-08-1332
  • Horlock C, Stott B, Dyson PJ, et al. The effects of trastuzumab on the CD4+CD25+FoxP3+ and CD4+IL17A+ T-cell axis in patients with breast cancer. Br J Cancer. 2009;100(7):1061–1067. doi:10.1038/sj.bjc.6604963
  • Steinberg SM, Zhang P, Malik BT, et al. BRAF inhibition alleviates immune suppression in murine autochthonous melanoma. Cancer Immunol Res. 2014;2(11):1044–1050. doi:10.1158/2326-6066.CIR-14-0074
  • Ghoreschi K, Laurence A, O’Shea JJ. Janus kinases in immune cell signaling. Immunol Rev. 2009;228(1):273–287. doi:10.1111/j.1600-065X.2008.00754.x
  • Iamsawat S, Daenthanasanmak A, Voss JH, et al. Stabilization of Foxp3 by targeting JAK2 enhances efficacy of CD8 induced regulatory T cells in the prevention of graft-versus-host disease. J Immunol. 2018;201(9):2812–2823. doi:10.4049/jimmunol.1800793
  • Barbui T, Carobbio A, Finazzi G, et al. Inflammation and thrombosis in essential thrombocythemia and polycythemia vera: different role of C-reactive protein and pentraxin 3. Haematologica. 2011;96(2):315–318. doi:10.3324/haematol.2010.031070
  • Hasselbalch HC, Bjørn ME. MPNs as inflammatory diseases: the evidence, consequences, and perspectives. Mediators Inflamm. 2015;2015:102476. doi:10.1155/2015/102476
  • Fragoulis GE, McInnes IB, Siebert S. JAK-inhibitors. New players in the field of immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology (Oxford). 2019;58(Suppl1):i43–i54. doi:10.1093/rheumatology/key276
  • Pei H, He L, Shao M, et al. Discovery of a highly selective JAK3 inhibitor for the treatment of rheumatoid arthritis. Sci Rep. 2018;8(1):5273. doi:10.1038/s41598-018-23569-y
  • Fleischmann RM, Damjanov NS, Kivitz AJ, Legedza A, Hoock T, Kinnman N. A randomized, double-blind, placebo-controlled, twelve-week, dose-ranging study of decernotinib, an oral selective JAK-3 inhibitor, as monotherapy in patients with active rheumatoid arthritis. Arthritis Rheumatol. 2015;67(2):334–343. doi:10.1002/art.38949
  • Shimoda K, Kato K, Aoki K, et al. Tyk2 plays a restricted role in IFN alpha signaling, although it is required for IL-12-mediated T cell function. Immunity. 2000;13(4):561–571. doi:10.1016/s1074-7613(00)00055-8
  • Ishizaki M, Akimoto T, Muromoto R, et al. Involvement of tyrosine kinase-2 in both the IL-12/Th1 and IL-23/Th17 axes in vivo. J Immunol. 2011;187(1):181–189. doi:10.4049/jimmunol.1003244
  • Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK-STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects [published correction appears in drugs.;77 (8):939][published correction appears in drugs. 2017 Jun 12]. Drugs. 2017;77(5):521–546. doi:10.1007/s40265-017-0701-9
  • Rother N. Disturbed T cell signaling and altered Th17 and regulatory T cell subsets in the pathogenesis of systemic lupus erythematosus. Front Immunol. 2015;6:610. doi:10.3389/fimmu.2015.00610
  • Bussel J, Arnold DM, Grossbard E, et al. Fostamatinib for the treatment of adult persistent and chronic immune thrombocytopenia: results of two Phase 3, randomized, placebo-controlled trials. Am J Hematol. 2018;93(7):921–930. doi:10.1002/ajh.25125
  • Newland A, Lee EJ, McDonald V, Bussel JB. Fostamatinib for persistent/chronic adult immune thrombocytopenia. Immunotherapy. 2018;10(1):9–25. doi:10.2217/imt-2017-0097
  • Keohane C, Kordasti S, Seidl T, et al. JAK inhibition induces silencing of T Helper cytokine secretion and a profound reduction in T regulatory cells. Br J Haematol. 2015;171(1):60–73. doi:10.1111/bjh.13519
  • Sewgobind VD, Quaedackers ME, van der Laan LJ, et al. The Jak inhibitor CP-690550 preserves the function of CD4CD25FoxP3 regulatory T cells and inhibits effector T cells. Am J Transplant. 2010;10(8):1785–1795. doi:10.1111/j.1600-6143.2010.03200.x
  • Meyer A, Wittekind PS, Kotschenreuther K, et al. Regulatory T cell frequencies in patients with rheumatoid arthritis are increased by conventional and biological DMARDs but not by JAK inhibitors. Ann Rheum Dis. 2019:annrheumdis-2019-216576. doi:10.1136/annrheumdis-2019-216576.
  • Alzahrani KS, Nadeem A, Ahmad SF, et al. Inhibition of spleen tyrosine kinase attenuates psoriasis-like inflammation in mice through blockade of dendritic cell-Th17 inflammation axis. Biomed Pharmacother. 2019;111:347–358. doi:10.1016/j.biopha.2018.12.060
  • Park JS, Lee J, Lim MA, et al. JAK2-STAT3 blockade by AG490 suppresses autoimmune arthritis in mice via reciprocal regulation of regulatory T cells and Th17 cells. J Immunol. 2014;192(9):4417–4424. doi:10.4049/jimmunol.1300514
  • Tanaka Y, McInnes IB, Taylor PC, et al. Characterization and changes of lymphocyte subsets in baricitinib-treated patients with rheumatoid arthritis: an integrated analysis. Arthritis Rheumatol. 2018;70(12):1923–1932. doi:10.1002/art.40680
  • Jasiecka-Mikolajczyk A, Socha P, Jaroszewski JJ, Maslanka T. Oclacitinib induces Foxp3 expression in CD4+ T cells under activation conditions. J Immunol. 2020;204(1 Supplement):92.
  • Betts BC, Bastian D, Iamsawat S, et al. Targeting JAK2 reduces GVHD and xenograft rejection through regulation of T cell differentiation. Proc Natl Acad Sci U S A. 2018;115(7):1582–1587. doi:10.1073/pnas.1712452115
  • Spoerl S, Mathew NR, Bscheider M, et al. Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease. Blood. 2014;123(24):3832–3842. doi:10.1182/blood-2013-12-543736
  • Parampalli Yajnanarayana S, Stübig T, Cornez I, et al. JAK1/2 inhibition impairs T cell function in vitro and in patients with myeloproliferative neoplasms. Br J Haematol. 2015;169(6):824–833. doi:10.1111/bjh.13373
  • Massa M, Rosti V, Campanelli R, Fois G, Barosi G. Rapid and long-lasting decrease of T-regulatory cells in patients with myelofibrosis treated with ruxolitinib. Leukemia. 2014;28(2):449–451. doi:10.1038/leu.2013.296
  • Virtanen T, Haikarainen T, Raivola J, Silvennoinen O. Selective JAKinibs: prospects in inflammatory and autoimmune diseases. BioDrugs. 2019;33(1):15–32. doi:10.1007/s40259-019-00333-w
  • Sigismund S, Avanzato D, Lanzetti L. Emerging functions of the EGFR in cancer. Mol Oncol. 2018;12(1):3–20. doi:10.1002/1878-0261.12155
  • Zaiss DM, van Loosdregt J, Gorlani A, et al. Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor. Immunity. 2013;38(2):275–284. doi:10.1016/j.immuni.2012.09.023
  • Apte RS, Chen DS, Ferrara N. VEGF in signaling and disease: beyond discovery and development. Cell. 2019;176(6):1248–1264. doi:10.1016/j.cell.2019.01.021
  • Zhu P, Hu C, Hui K, Jiang X. The role and significance of VEGFR2+ regulatory T cells in tumor immunity. Onco Targets Ther. 2017;10:4315–4319. doi:10.2147/OTT.S142085
  • Farooqi AA, Siddik ZH. Platelet-derived growth factor (PDGF) signalling in cancer: rapidly emerging signalling landscape. Cell Biochem Funct. 2015;33(5):257–265. doi:10.1002/cbf.3120
  • Agrawal S, Ganguly S, Hajian P, Cao JN, Agrawal A. PDGF upregulates CLEC-2 to induce T regulatory cells. Oncotarget. 2015;6(30):28621–28632. doi:10.18632/oncotarget.5765
  • Croce L, Coperchini F, Magri F, Chiovato L, Rotondi M. The multifaceted anti-cancer effects of BRAF-inhibitors. Oncotarget. 2019;10(61):6623–6640. doi:10.18632/oncotarget.27304
  • Shabaneh TB, Molodtsov AK, Steinberg SM, et al. Oncogenic BRAFV600E governs regulatory T-cell recruitment during melanoma tumorigenesis. Cancer Res. 2018;78(17):5038–5049. doi:10.1158/0008-5472.CAN-18-0365
  • Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949–954. doi:10.1038/nature00766
  • Kang ZJ, Liu YF, Xu LZ, et al. The Philadelphia chromosome in leukemogenesis. Chin J Cancer. 2016;35:48. doi:10.1186/s40880-016-0108-0
  • Manlove LS, Berquam-Vrieze KE, Pauken KE, Williams RT, Jenkins MK, Farrar MA. Adaptive immunity to leukemia is inhibited by cross-reactive induced regulatory T cells. J Immunol. 2015;195(8):4028–4037. doi:10.4049/jimmunol.1501291
  • 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/nri2889
  • Cai Y, Fleming C, Yan J. New insights of T cells in the pathogenesis of psoriasis. Cell Mol Immunol. 2012;9(4):302–309. doi:10.1038/cmi.2012.15
  • Federici M, Giustizieri ML, Scarponi C, Girolomoni G, Albanesi C. Impaired IFN-gamma-dependent inflammatory responses in human keratinocytes overexpressing the suppressor of cytokine signaling 1. J Immunol. 2002;169(1):434–442. doi:10.4049/jimmunol.169.1.434
  • Morelli M, Scarponi C, Mercurio L, et al. Selective immunomodulation of inflammatory pathways in keratinocytes by the janus kinase (JAK) inhibitor tofacitinib: implications for the employment of JAK-targeting drugs in psoriasis. J Immunol Res. 2018;2018:7897263. doi:10.1155/2018/7897263
  • Jones DS, Jenney AP, Swantek JL, Burke JM, Lauffenburger DA, Sorger PK. Profiling drugs for rheumatoid arthritis that inhibit synovial fibroblast activation. Nat Chem Biol. 2017;13(1):38–45. doi:10.1038/nchembio.2211
  • Vainchenker W, Leroy E, Gilles L, Marty C, Plo I, Constantinescu SN. JAK inhibitors for the treatment of myeloproliferative neoplasms and other disorders. F1000Res. 2018;7:82. doi:10.12688/f1000research.13167.1
  • Liotta F, Gacci M, Frosali F, et al. Frequency of regulatory T cells in peripheral blood and in tumour-infiltrating lymphocytes correlates with poor prognosis in renal cell carcinoma. BJU Int. 2011;107(9):1500–1506. doi:10.1111/j.1464-410X.2010.09555.x
  • Medicines.org.uk; Pfizer Limited; 2021. Inlyta 5 mg film coated tablet; Summary of Product Characteristics. Available from: https://www.medicines.org.uk/emc/product/7948. Accessed 4 March 2021. 
  • Medicines.org.uk; Roche Products Limited; 2021. Avastin 25mg/ml concentrate for solution for infusion; Summary of Product Characteristics. Available from: https://www.medicines.org.uk/emc/product/3885. Accessed 4 March 2021.
  • Wijarnpreecha K, Nissaisorakarn P, Sornprom S, et al. Hepatitis C infection and renal cell carcinoma: a systematic review and meta-analysis. World J Gastrointest Pathophysiol. 2016;7(4):314–319. doi:10.4291/wjgp.v7.i4.314
  • Farhadi A, Behzad-Behbahani A, Geramizadeh B, et al. Presence of oncogenic viruses (EBV, HHV-6, BKV and JCV) DNA sequences in renal cell carcinoma. Int J Infect Dis. 2012;16(1):e80. doi:10.1016/j.ijid.2012.05.193
  • Shimakage M, Kawahara K, Harada S, Sasagawa T, Shinka T, Oka T. Expression of Epstein-Barr virus in renal cell carcinoma. Oncol Rep. 2007;18(1):41–46.
  • Heidegger I, Borena W, Pichler R. The role of human papilloma virus in urological malignancies. Anticancer Res. 2015;35(5):2513–2519.
  • Liu C, Wu S, Meng X, et al. Predictive value of peripheral regulatory T cells in non-small cell lung cancer patients undergoing radiotherapy. Oncotarget. 2017;8(26):43427–43438. doi:10.18632/oncotarget.15238
  • Shimizu K, Nakata M, Hirami Y, Yukawa T, Maeda A, Tanemoto K. Tumor-infiltrating Foxp3+ regulatory T cells are correlated with cyclooxygenase-2 expression and are associated with recurrence in resected non-small cell lung cancer. J Thorac Oncol. 2010;5(5):585–590. doi:10.1097/JTO.0b013e3181d60fd7
  • Zhu QG, Zhang SM, Ding XX, He B, Zhang HQ. Driver genes in non-small cell lung cancer: characteristics, detection methods, and targeted therapies. Oncotarget. 2017;8(34):57680–57692. doi:10.18632/oncotarget.17016
  • Liu Y, O’Brien JL, Ajami NJ, et al. Lung tissue microbial profile in lung cancer is distinct from emphysema. Am J Cancer Res. 2018;8(9):1775–1787.
  • O’Mahony C, Scully P, O’Mahony D, et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog. 2008;4(8):e1000112. doi:10.1371/journal.ppat.1000112
  • Mertens J, Fabri M, Zingarelli A, et al. Streptococcus pneumoniae serotype 1 capsular polysaccharide induces CD8CD28 regulatory T lymphocytes by TCR crosslinking. PLoS Pathog. 2009;5(9):e1000596. doi:10.1371/journal.ppat.1000596
  • Larsen JM. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology. 2017;151(4):363–374. doi:10.1111/imm.12760
  • Mangalam A, Shahi SK, Luckey D, et al. Human gut-derived commensal bacteria suppress CNS inflammatory and demyelinating disease. Cell Rep. 2017;20(6):1269–1277. doi:10.1016/j.celrep.2017.07.031
  • Robinson LA, Jaing CJ, Pierce Campbell C, et al. Molecular evidence of viral DNA in non-small cell lung cancer and non-neoplastic lung. Br J Cancer. 2016;115(4):497–504. doi:10.1038/bjc.2016.213
  • Kheir F, Zhao M, Strong MJ, et al. Detection of Epstein-Barr virus infection in non-small cell lung cancer. Cancers (Basel). 2019;11(6):759. doi:10.3390/cancers11060759
  • Landtblom AR, Andersson TM, Dickman PW, et al. Risk of infections in patients with myeloproliferative neoplasms-a population-based cohort study of 8363 patients [published online ahead of print, 2020 Jun 16]. Leukemia. 2020. doi:10.1038/s41375-020-0909-7.
  • Ruan S, Tate C, Lee JJ, Ritter T, Kolls JK, Shellito JE. Local delivery of the viral interleukin-10 gene suppresses tissue inflammation in murine Pneumocystis carinii infection. Infect Immun. 2002;70(11):6107–6113. doi:10.1128/iai.70.11.6107-6113.2002
  • Wright TW, Gigliotti F, Finkelstein JN, McBride JT, An CL, Harmsen AG. Immune-mediated inflammation directly impairs pulmonary function, contributing to the pathogenesis of Pneumocystis carinii pneumonia. J Clin Invest. 1999;104(9):1307–1317. doi:10.1172/JCI6688
  • Shrivastava S, TrehanPati N, Patra S, et al. Increased regulatory T cells and impaired functions of circulating CD8 T lymphocytes is associated with viral persistence in Hepatitis B virus-positive newborns. J Viral Hepat. 2013;20(8):582–591. doi:10.1111/jvh.12078
  • Kolata JB, Kühbandner I, Link C, et al. The fall of a dogma? Unexpected high T-cell memory response to Staphylococcus aureus in humans. J Infect Dis. 2015;212(5):830–838. doi:10.1093/infdis/jiv128
  • Frodermann V, Chau TA, Sayedyahossein S, Toth JM, Heinrichs DE, Madrenas J. A modulatory interleukin-10 response to staphylococcal peptidoglycan prevents Th1/Th17 adaptive immunity to Staphylococcus aureus. J Infect Dis. 2011;204(2):253–262. doi:10.1093/infdis/jir276
  • Ferraro A, Buonocore SM, Auquier P, et al. Role and plasticity of Th1 and Th17 responses in immunity to Staphylococcus aureus. Hum Vaccin Immunother. 2019;15(12):2980–2992. doi:10.1080/21645515.2019.1613126
  • Rose WE, Shukla SK, Berti AD, et al. Increased endovascular staphylococcus aureus inoculum is the link between elevated serum interleukin 10 concentrations and mortality in patients with bacteremia [published correction appears in clin infect dis. 2017 Oct 15;65(8):1431–1433]. Clin Infect Dis. 2017;64(10):1406–1412. doi:10.1093/cid/cix157
  • Zhang J, Zhu Z, Zuo X, et al. The role of NTHi colonization and infection in the pathogenesis of neutrophilic asthma. Respir Res. 2020;21(1):170. doi:10.1186/s12931-020-01438-5
  • Essilfie AT, Simpson JL, Horvat JC, et al. Haemophilus influenzae infection drives IL-17-mediated neutrophilic allergic airways disease. PLoS Pathog. 2011;7(10):e1002244. doi:10.1371/journal.ppat.1002244
  • Guan X, Lu Y, Wang G, et al. The role of regulatory T cell in nontypeable haemophilus influenzae-induced acute exacerbation of chronic obstructive pulmonary disease. Mediators Inflamm. 2018;2018:8387150. doi:10.1155/2018/8387150
  • Kim MS, Kim DJ, Na CH, Shin BSA. Study of the changes of T helper 17 cells and regulatory T cells in herpes zoster. Ann Dermatol. 2017;29(5):578–585. doi:10.5021/ad.2017.29.5.578
  • Betts RJ, Prabhu N, Ho AW, et al. Influenza A virus infection results in a robust, antigen-responsive, and widely disseminated Foxp3+ regulatory T cell response. J Virol. 2012;86(5):2817–2825. doi:10.1128/JVI.05685-11
  • Kudva A, Scheller EV, Robinson KM, et al. Influenza A inhibits Th17-mediated host defense against bacterial pneumonia in mice. J Immunol. 2011;186(3):1666–1674. doi:10.4049/jimmunol.1002194
  • Brereton CF, Sutton CE, Ross PJ, et al. Escherichia coli heat-labile enterotoxin promotes protective Th17 responses against infection by driving innate IL-1 and IL-23 production. J Immunol. 2011;186(10):5896–5906. doi:10.4049/jimmunol.1003789
  • Luo J, Zhang M, Yan B, Zhang K, Chen M, Deng S. Imbalance of Th17 and Treg in peripheral blood mononuclear cells of active tuberculosis patients. Braz J Infect Dis. 2017;21(2):155–161. doi:10.1016/j.bjid.2016.10.011
  • Nikitina IY, Panteleev AV, Kosmiadi GA, et al. Th1, Th17, and Th1Th17 lymphocytes during tuberculosis: th1 lymphocytes predominate and appear as low-differentiated CXCR3+CCR6+ cells in the blood and highly differentiated CXCR3±CCR6- cells in the lungs. J Immunol. 2018;200(6):2090–2103. doi:10.4049/jimmunol.1701424
  • Pai M, Denkinger CM, Kik SV, et al. Gamma interferon release assays for detection of Mycobacterium tuberculosis infection. Clin Microbiol Rev. 2014;27(1):3–20. doi:10.1128/CMR.00034-13
  • Li Q, Li J, Tian J, et al. IL-17 and IFN-γ production in peripheral blood following BCG vaccination and Mycobacterium tuberculosis infection in human. Eur Rev Med Pharmacol Sci. 2012;16(14):2029–2036.
  • Aung H, Toossi Z, McKenna SM, et al. Expression of transforming growth factor-beta but not tumor necrosis factor-alpha, interferon-gamma, and interleukin-4 in granulomatous lung lesions in tuberculosis. Tuber Lung Dis. 2000;80(2):61–67. doi:10.1054/tuld.2000.0235
  • Wu M, Aung H, Hirsch CS, Toossi Z. Inhibition of Mycobacterium tuberculosis-induced signalling by transforming growth factor-β in human mononuclear phagocytes. Scand J Immunol. 2012;75(3):301–304. doi:10.1111/j.1365-3083.2011.02668.x
  • 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.2014.08.010
  • Bentires-Alj M, Paez JG, David FS, et al. Activating mutations of the noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia. Cancer Res. 2004;64(24):8816–8820. doi:10.1158/0008-5472.CAN-04-1923
  • Lee IO, Kim JH, Choi YJ, et al. Helicobacter pylori CagA phosphorylation status determines the gp130-activated SHP2/ERK and JAK/STAT signal transduction pathways in gastric epithelial cells. J Biol Chem. 2010;285(21):16042–16050. doi:10.1074/jbc.M110.111054
  • Neumann WL, Coss E, Rugge M, Genta RM. Autoimmune atrophic gastritis–pathogenesis, pathology and management. Nat Rev Gastroenterol Hepatol. 2013;10(9):529–541. doi:10.1038/nrgastro.2013.101
  • Shi WJ, Liu W, Zhou XY, Ye F, Zhang GX. Associations of Helicobacter pylori infection and cytotoxin-associated gene A status with autoimmune thyroid diseases: a meta-analysis. Thyroid. 2013;23(10):1294–1300. doi:10.1089/thy.2012.0630
  • Smyk DS, Koutsoumpas AL, Mytilinaiou MG, Rigopoulou EI, Sakkas LI, Bogdanos DP. Helicobacter pylori and autoimmune disease: cause or bystander. World J Gastroenterol. 2014;20(3):613–629. doi:10.3748/wjg.v20.i3.613
  • Miller WE, Mosialos G, Kieff E, Raab-Traub N. Epstein-Barr virus LMP1 induction of the epidermal growth factor receptor is mediated through a TRAF signaling pathway distinct from NF-kappaB activation. J Virol. 1997;71(1):586–594. doi:10.1128/JVI.71.1.586-594.1997
  • Dawson CW, Laverick L, Morris MA, Tramoutanis G, Young LS. Epstein-Barr virus-encoded LMP1 regulates epithelial cell motility and invasion via the ERK-MAPK pathway. J Virol. 2008;82(7):3654–3664. doi:10.1128/JVI.01888-07
  • Dawson CW, Tramountanis G, Eliopoulos AG, Young LS. Epstein-Barr virus latent membrane protein 1 (LMP1) activates the phosphatidylinositol 3-kinase/Akt pathway to promote cell survival and induce actin filament remodeling. J Biol Chem. 2003;278(6):3694–3704. doi:10.1074/jbc.M209840200
  • Vaysberg M, Lambert SL, Krams SM, Martinez OM. Activation of the JAK/STAT pathway in Epstein Barr virus+-associated posttransplant lymphoproliferative disease: role of interferon-gamma. Am J Transplant. 2009;9(10):2292–2302. doi:10.1111/j.1600-6143.2009.02781.x
  • Ando S, Kawada JI, Watanabe T, et al. Tofacitinib induces G1 cell-cycle arrest and inhibits tumor growth in Epstein-Barr virus-associated T and natural killer cell lymphoma cells. Oncotarget. 2016;7(47):76793–76805. doi:10.18632/oncotarget.12529
  • Fujiwara S, Takei M. Epstein‐Barr virus and autoimmune diseases. Clin Exp Neuroimmunol. 2015;6:38–48. doi:10.1111/cen3.12263
  • 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(1):15179. doi:10.1038/srep15179
  • Ueki IF, Min-Oo G, Kalinowski A, et al. Respiratory virus-induced EGFR activation suppresses IRF1-dependent interferon λ and antiviral defense in airway epithelium. J Exp Med. 2013;210(10):1929–1936. doi:10.1084/jem.20121401
  • Alkharsah KR. VEGF upregulation in viral infections and its possible therapeutic implications. Int J Mol Sci. 2018;19(6):1642. doi:10.3390/ijms19061642
  • Soroceanu L, Akhavan A, Cobbs CS. Platelet-derived growth factor-alpha receptor activation is required for human cytomegalovirus infection. Nature. 2008;455(7211):391–395. doi:10.1038/nature07209
  • DuShane JK, Maginnis MS. Human DNA virus exploitation of the MAPK-ERK cascade. Int J Mol Sci. 2019;20(14):3427. doi:10.3390/ijms20143427
  • Benn J, Schneider RJ. Hepatitis B virus HBx protein activates Ras-GTP complex formation and establishes a Ras, Raf, MAP kinase signaling cascade. Proc Natl Acad Sci U S A. 1994;91(22):10350–10354. doi:10.1073/pnas.91.22.10350
  • Klein NP, Schneider RJ. Activation of Src family kinases by hepatitis B virus HBx protein and coupled signaling to Ras. Mol Cell Biol. 1997;17(11):6427–6436. doi:10.1128/mcb.17.11.6427
  • Tran TTP, Eichholz K, Amelio P, et al. Humoral immune response to adenovirus induce tolerogenic bystander dendritic cells that promote generation of regulatory T cells. PLoS Pathog. 2018;14(8):e1007127. doi:10.1371/journal.ppat.1007127
  • Gaido CM, Granland C, Laing IA, et al. T-cell responses against rhinovirus species A and C in asthmatic and healthy children. Immun Inflamm Dis. 2018;6(1):143–153. doi:10.1002/iid3.206
  • Tovar-Salazar A, Weinberg A. Understanding the mechanism of action of cytomegalovirus-induced regulatory T cells. Virology. 2020;547:1–6. doi:10.1016/j.virol.2020.05.001
  • Jayaratne HE, Wijeratne D, Fernando S, et al. Regulatory T-cells in acute dengue viral infection. Immunology. 2018;154(1):89–97. doi:10.1111/imm.12863
  • Benson A, Murray S, Divakar P, et al. Microbial infection-induced expansion of effector T cells overcomes the suppressive effects of regulatory T cells via an IL-2 deprivation mechanism. J Immunol. 2012;188(2):800–810. doi:10.4049/jimmunol.1100769
  • Fleming SB. Viral inhibition of the IFN-induced JAK/STAT signalling pathway: development of live attenuated vaccines by mutation of viral-encoded IFN-antagonists. Vaccines (Basel). 2016;4(3):23. doi:10.3390/vaccines4030023
  • Kak G, Raza M, Tiwari BK. Interferon-gamma (IFN-γ): exploring its implications in infectious diseases. Biomol Concepts. 2018;9(1):64–79. doi:10.1515/bmc-2018-0007
  • Gerada C, Campbell TM, Kennedy JJ, et al. Manipulation of the innate immune response by varicella zoster virus. Front Immunol. 2020;11:1. doi:10.3389/fimmu.2020.00001
  • Beekhuizen H, van de Gevel JS. Gamma interferon confers resistance to infection with Staphylococcus aureus in human vascular endothelial cells by cooperative proinflammatory and enhanced intrinsic antibacterial activities. Infect Immun. 2007;75(12):5615–5626. doi:10.1128/IAI.00530-07
  • Norkina O, Dolganiuc A, Shapiro T, Kodys K, Mandrekar P, Szabo G. Acute alcohol activates STAT3, AP-1, and Sp-1 transcription factors via the family of Src kinases to promote IL-10 production in human monocytes. J Leukoc Biol. 2007;82(3):752–762. doi:10.1189/jlb.0207099
  • Psaltopoulou T, Sergentanis TN, Ntanasis-Stathopoulos I, Tzanninis IG, Tsilimigras DI, Dimopoulos MA. Alcohol consumption and risk of hematological malignancies: a meta-analysis of prospective studies. Int J Cancer. 2018;143(3):486–495. doi:10.1002/ijc.31330
  • Rasouli B, Ahlbom A, Andersson T, et al. Alcohol consumption is associated with reduced risk of type 2 diabetes and autoimmune diabetes in adults: results from the Nord-Trøndelag health study. Diabet Med. 2013;30(1):56–64. doi:10.1111/j.1464-5491.2012.03713.x
  • Zeng C, Shi X, Zhang B, et al. The imbalance of Th17/Th1/Tregs in patients with type 2 diabetes: relationship with metabolic factors and complications. J Mol Med (Berl). 2012;90(2):175–186. doi:10.1007/s00109-011-0816-5
  • Farag AGA, Samaka R, Elshafey EN, Shehata WA, El Sherbiny EG, Hammam MA. Immunohistochemical study of janus kinase 1/signal transducer and activator of transcription 3 in psoriasis vulgaris. Clin Cosmet Investig Dermatol. 2019;12:497–508. doi:10.2147/CCID.S202835
  • Gruber CN, Calis JJA, Buta S, et al. Complex autoinflammatory syndrome unveils fundamental principles of JAK1 kinase transcriptional and biochemical function. Immunity. 2020;53(3):672–684.e11. doi:10.1016/j.immuni.2020.07.006
  • Del Bel KL, Ragotte RJ, Saferali A, et al. JAK1 gain-of-function causes an autosomal dominant immune dysregulatory and hypereosinophilic syndrome. J Allergy Clin Immunol. 2017;139(6):2016–2020.e5. doi:10.1016/j.jaci.2016.12.957
  • Arulogun SO, Choong HL, Taylor D, et al. JAK1 somatic mutation in a myeloproliferative neoplasm. Haematologica. 2017;102(8):e324–e327. doi:10.3324/haematol.2017.170266
  • Albacker LA, Wu J, Smith P, et al. Loss of function JAK1 mutations occur at high frequency in cancers with microsatellite instability and are suggestive of immune evasion. PLoS One. 2017;12(11):e0176181. doi:10.1371/journal.pone.0176181
  • Ren Y, Zhang Y, Liu RZ, et al. JAK1 truncating mutations in gynecologic cancer define new role of cancer-associated protein tyrosine kinase aberrations. Sci Rep. 2013;3(1):3042. doi:10.1038/srep03042
  • Hu J, Wu X, Huang P, Teng F, Wang Y, Xue F. The proportion and prognostic significance of T-regulatory cells in patients with gynecological cancers: a systematic review and meta-analysis. J Cancer. 2020;11(11):3340–3348. doi:10.7150/jca.42472
  • Zhang X, Kelaria S, Kerstetter J, Wang J. The functional and prognostic implications of regulatory T cells in colorectal carcinoma. J Gastrointest Oncol. 2015;6(3):307–313. doi:10.3978/j.issn.2078-6891.2015.017
  • Liu X, Zhang Z, Zhao G. Recent advances in the study of regulatory T cells in gastric cancer. Int Immunopharmacol. 2019;73:560–567. doi:10.1016/j.intimp.2019.05.009
  • Dai J, Lu Y, Roca H, et al. Immune mediators in the tumor microenvironment of prostate cancer. Chin J Cancer. 2017;36(1):29. doi:10.1186/s40880-017-0198-3
  • Xu P, Fan W, Zhang Z, et al. The clinicopathological and prognostic implications of FoxP3+ regulatory T cells in patients with colorectal cancer: a meta-analysis. Front Physiol. 2017;8:950. doi:10.3389/fphys.2017.00950
  • Paniagua RT, Sharpe O, Ho PP, et al. Selective tyrosine kinase inhibition by imatinib mesylate for the treatment of autoimmune arthritis. J Clin Invest. 2006;116(10):2633–2642. doi:10.1172/JCI28546
  • Eklund KK, Joensuu H. Treatment of rheumatoid arthritis with imatinib mesylate: clinical improvement in three refractory cases [published correction appears in Ann Med. 2003;35(8):640]. Ann Med. 2003;35(5):362–367. doi:10.1080/07853890310001339
  • Eklund KK, Remitz A, Kautiainen H, Reitamo S, Leirisalo-Repo M. Three months treatment of active spondyloarthritis with imatinib mesylate: an open-label pilot study with six patients. Rheumatology (Oxford). 2006;45(12):1573–1575. doi:10.1093/rheumatology/kel365
  • Kuang YH, Lu Y, Liu YK, et al. Topical Sunitinib ointment alleviates Psoriasis-like inflammation by inhibiting the proliferation and apoptosis of keratinocytes. Eur J Pharmacol. 2018;824:57–63. doi:10.1016/j.ejphar.2018.01.048
  • Ozgen M, Koca SS, Karatas A, et al. Lapatinib ameliorates experimental arthritis in rats. Inflammation. 2015;38(1):252–259. doi:10.1007/s10753-014-0028-6
  • Gore J, Craven KE, Wilson JL, et al. TCGA data and patient-derived orthotopic xenografts highlight pancreatic cancer-associated angiogenesis. Oncotarget. 2015;6(10):7504–7521. doi:10.18632/oncotarget.3233
  • Koblish HK, Hansbury M, Wang L-CS, et al. Novel immunotherapeutic activity of JAK and PI3Kδ inhibitors in a model of pancreatic cancer [abstract]. Cancer Res. 2015;75:1336. doi:10.1158/1538-7445.am2015-1336
  • Zhu Z, Aref AR, Cohoon TJ, et al. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit. Cancer Discov. 2014;4(4):452–465. doi:10.1158/2159-8290.CD-13-0646
  • Barbie DA, Spira A, Kelly K, et al. Phase 1B study of momelotinib combined with trametinib in metastatic, kirsten rat sarcoma viral oncogene homolog-mutated non-small-cell lung cancer after platinum-based chemotherapy treatment failure. Clin Lung Cancer. 2018;19(6):e853–e859. doi:10.1016/j.cllc.2018.07.004
  • Ng K, Hendifar A, Starodub A, et al. Phase 1 dose-escalation study of momelotinib, a Janus kinase 1/2 inhibitor, combined with gemcitabine and nab-paclitaxel in patients with previously untreated metastatic pancreatic ductal adenocarcinoma. Invest New Drugs. 2019;37(1):159–165. doi:10.1007/s10637-018-0650-5
  • Fogelman D, Cubillo A, García-Alfonso P, et al. Randomized, double-blind, phase two study of ruxolitinib plus regorafenib in patients with relapsed/refractory metastatic colorectal cancer. Cancer Med. 2018;7(11):5382–5393. doi:10.1002/cam4.1703
  • Hurwitz HI, Uppal N, Wagner SA, et al. Randomized, double-blind, phase II study of ruxolitinib or placebo in combination with capecitabine in patients with metastatic pancreatic cancer for whom therapy with gemcitabine has failed. J Clin Oncol. 2015;33(34):4039–4047. doi:10.1200/JCO.2015.61.4578
  • Hurwitz H, Van Cutsem E, Bendell J, et al. Ruxolitinib + capecitabine in advanced/metastatic pancreatic cancer after disease progression/intolerance to first-line therapy: JANUS 1 and 2 randomized phase III studies. Invest New Drugs. 2018;36(4):683–695. doi:10.1007/s10637-018-0580-2