메뉴 건너뛰기




Volumn 19, Issue 4, 2018, Pages 326-344

Immune therapy in sepsis: Are we ready to try again?

Author keywords

adjuvant immune therapy; immune biomarker; immune suppression; Sepsis; sepsis immune pathogenesis

Indexed keywords

ALBUMIN; B7 ANTIGEN; BIOLOGICAL MARKER; C REACTIVE PROTEIN; CD135 ANTIGEN; CD28 ANTIGEN; CD74 ANTIGEN; CD86 ANTIGEN; CREATININE; CYTOKINE; CYTOTOXIC T LYMPHOCYTE ANTIGEN 4; GAMMA INTERFERON; GRANULOCYTE COLONY STIMULATING FACTOR; GRANULOCYTE MACROPHAGE COLONY STIMULATING FACTOR; HLA DR ANTIGEN; IMMUNOGLOBULIN; INTERLEUKIN 1; INTERLEUKIN 6; MACROPHAGE INFLAMMATORY PROTEIN 1BETA; MAJOR HISTOCOMPATIBILITY ANTIGEN CLASS 2; NIVOLUMAB; PLASMA PROTEIN; PROCALCITONIN; PROGRAMMED DEATH 1 LIGAND 1; PROGRAMMED DEATH 1 RECEPTOR; TRANSFORMING GROWTH FACTOR BETA1; TRIGGERING RECEPTOR EXPRESSED ON MYELOID CELLS 1; TUMOR NECROSIS FACTOR; TUMOR NECROSIS FACTOR RECEPTOR; UNINDEXED DRUG;

EID: 85045032633     PISSN: 17511437     EISSN: None     Source Type: Journal    
DOI: 10.1177/1751143718765407     Document Type: Review
Times cited : (52)

References (170)
  • 1
    • 84959273475 scopus 로고    scopus 로고
    • The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3)
    • Singer M, Deutschman CS, Seymour CW, The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016; 315: 801–810
    • (2016) JAMA , vol.315 , pp. 801-810
    • Singer, M.1    Deutschman, C.S.2    Seymour, C.W.3
  • 2
    • 31344461826 scopus 로고    scopus 로고
    • Sepsis in European intensive care units: results of the SOAP study
    • Vincent J-L, Sakr Y, Sprung CL, Sepsis in European intensive care units: results of the SOAP study. Crit Care Med 2006; 34: 344–353
    • (2006) Crit Care Med , vol.34 , pp. 344-353
    • Vincent, J.-L.1    Sakr, Y.2    Sprung, C.L.3
  • 3
    • 85045954751 scopus 로고    scopus 로고
    • Nutbeam T, Birmingham: United Kingdom Sepsis Trust, (2017, accessed 9 March 2018)
    • Daniels R (ed.), Nutbeam T (ed.). The Sepsis Manual. Birmingham: United Kingdom Sepsis Trust; 2017. Available from https://sepsistrust.org/wp-content/uploads/2018/02/Sepsis_Manual_2017_web_download.pdf (2017, accessed 9 March 2018)
    • (2017) The Sepsis Manual
    • Daniels, R.1
  • 4
    • 84899044664 scopus 로고    scopus 로고
    • Is boosting the immune system in sepsis appropriate?
    • Cavaillon JM, Eisen D, Annane D. Is boosting the immune system in sepsis appropriate? Crit Care 2014; 18: 216
    • (2014) Crit Care , vol.18 , pp. 216
    • Cavaillon, J.M.1    Eisen, D.2    Annane, D.3
  • 5
    • 84897377065 scopus 로고    scopus 로고
    • Why have clinical trials in sepsis failed?
    • Marshall JC. Why have clinical trials in sepsis failed? Trends Mol Med 2014; 20: 195–203
    • (2014) Trends Mol Med , vol.20 , pp. 195-203
    • Marshall, J.C.1
  • 6
    • 84891645973 scopus 로고    scopus 로고
    • The changing immune system in sepsis: is individualized immuno-modulatory therapy the answer?
    • Boomer JS, Green JM, Hotchkiss RS. The changing immune system in sepsis: is individualized immuno-modulatory therapy the answer? Virulence 2014; 5: 45–56
    • (2014) Virulence , vol.5 , pp. 45-56
    • Boomer, J.S.1    Green, J.M.2    Hotchkiss, R.S.3
  • 7
    • 84873339299 scopus 로고    scopus 로고
    • Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012
    • Levy MM, Rhodes A, Annane D, Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 2013; 41: 580–637
    • (2013) Crit Care Med , vol.41 , pp. 580-637
    • Levy, M.M.1    Rhodes, A.2    Annane, D.3
  • 9
    • 79751531129 scopus 로고    scopus 로고
    • Granulocyte-colony stimulating factor (G-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF) for sepsis: a meta-analysis
    • Bo L, Wang F, Zhu J, Granulocyte-colony stimulating factor (G-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF) for sepsis: a meta-analysis. Crit Care 2011; 15: R58
    • (2011) Crit Care , vol.15 , pp. R58
    • Bo, L.1    Wang, F.2    Zhu, J.3
  • 10
    • 34249943678 scopus 로고    scopus 로고
    • The advent of the cytokine storm
    • Clark IA. The advent of the cytokine storm. Immunol Cell Biol 2007; 85: 271–273
    • (2007) Immunol Cell Biol , vol.85 , pp. 271-273
    • Clark, I.A.1
  • 11
    • 0029903114 scopus 로고    scopus 로고
    • Sir Isaac Newton, sepsis, SIRS, and CARS
    • Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS. Crit Care Med 1996; 24: 1125–1128
    • (1996) Crit Care Med , vol.24 , pp. 1125-1128
    • Bone, R.C.1
  • 12
    • 84896671549 scopus 로고    scopus 로고
    • Immunological characterization of compensatory anti-inflammatory response syndrome in patients with severe sepsis
    • Gomez HG, Gonzalez SM, London JM, et al. Immunological characterization of compensatory anti-inflammatory response syndrome in patients with severe sepsis. Crit Care Med 2014; 42: 771–780
    • (2014) Crit Care Med , vol.42 , pp. 771-780
    • Gomez, H.G.1    Gonzalez, S.M.2    London, J.M.3
  • 13
    • 0036893263 scopus 로고    scopus 로고
    • Septic Plasma”: an immunosuppressive milieu
    • Cavaillon JM. “Septic Plasma”: an immunosuppressive milieu. Am J Respir Crit Care Med 2002; 166: 1417–1418
    • (2002) Am J Respir Crit Care Med , vol.166 , pp. 1417-1418
    • Cavaillon, J.M.1
  • 14
    • 66749164669 scopus 로고    scopus 로고
    • The sepsis seesaw: tilting toward immunosuppression
    • Hotchkiss RS, McDunn JE, Ferguson TA. The sepsis seesaw: tilting toward immunosuppression. Nat Med 2009; 15: 496–497
    • (2009) Nat Med , vol.15 , pp. 496-497
    • Hotchkiss, R.S.1    McDunn, J.E.2    Ferguson, T.A.3
  • 15
    • 84963568655 scopus 로고    scopus 로고
    • Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study
    • Davenport EE, Burnham KL, Radhakrishnan J, Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study. Lancet Respir Med 2016; 4: 259–271
    • (2016) Lancet Respir Med , vol.4 , pp. 259-271
    • Davenport, E.E.1    Burnham, K.L.2    Radhakrishnan, J.3
  • 16
    • 78650624395 scopus 로고    scopus 로고
    • Genome-wide transcription profiling of human sepsis: a systematic review
    • Tang BM, Huang SJ, McLean AS. Genome-wide transcription profiling of human sepsis: a systematic review. Crit Care 2010; 14: R237
    • (2010) Crit Care , vol.14 , pp. R237
    • Tang, B.M.1    Huang, S.J.2    McLean, A.S.3
  • 17
    • 27644597252 scopus 로고    scopus 로고
    • Reprogramming of circulatory cells in sepsis and SIRS
    • Cavaillon JM, Adrie C, Fitting C, Reprogramming of circulatory cells in sepsis and SIRS. J Endotoxin Res 2005; 11: 311–320
    • (2005) J Endotoxin Res , vol.11 , pp. 311-320
    • Cavaillon, J.M.1    Adrie, C.2    Fitting, C.3
  • 18
    • 0029823055 scopus 로고    scopus 로고
    • Monocyte deactivation–rationale for a new therapeutic strategy in sepsis
    • Volk H-D, Reinke P, Krausch D, Monocyte deactivation–rationale for a new therapeutic strategy in sepsis. Intensive Care Med 1996; 22(Suppl 4): S474–S481
    • (1996) Intensive Care Med , vol.22 , pp. S474-S481
    • Volk, H.-D.1    Reinke, P.2    Krausch, D.3
  • 19
    • 84963768824 scopus 로고    scopus 로고
    • Sepsis-induced long-term immune paralysis–results of a descriptive, explorative study
    • Arens C, Bajwa SA, Koch C, Sepsis-induced long-term immune paralysis–results of a descriptive, explorative study. Crit Care 2016; 20: 93
    • (2016) Crit Care , vol.20 , pp. 93
    • Arens, C.1    Bajwa, S.A.2    Koch, C.3
  • 20
    • 84888325490 scopus 로고    scopus 로고
    • Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy
    • Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol 2013; 13: 862–874
    • (2013) Nat Rev Immunol , vol.13 , pp. 862-874
    • Hotchkiss, R.S.1    Monneret, G.2    Payen, D.3
  • 21
    • 84255194001 scopus 로고    scopus 로고
    • Immunosuppression in patients who die of sepsis and multiple organ failure
    • Boomer JS, Faist E, Wichmann M, Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 2011; 306: 2594–2605
    • (2011) JAMA , vol.306 , pp. 2594-2605
    • Boomer, J.S.1    Faist, E.2    Wichmann, M.3
  • 22
    • 66349131288 scopus 로고    scopus 로고
    • Macroscopic postmortem findings in 235 surgical intensive care patients with sepsis
    • Torgersen C, Moser P, Luckner G, Macroscopic postmortem findings in 235 surgical intensive care patients with sepsis. Anesth Analg 2009; 108: 1817–1841
    • (2009) Anesth Analg , vol.108 , pp. 1817-1841
    • Torgersen, C.1    Moser, P.2    Luckner, G.3
  • 23
    • 84864545901 scopus 로고    scopus 로고
    • Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care
    • Gentile LF, Cuenca AG, Efron PA, Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J Trauma Acute Care Surg 2012; 72: 1491–1501
    • (2012) J Trauma Acute Care Surg , vol.72 , pp. 1491-1501
    • Gentile, L.F.1    Cuenca, A.G.2    Efron, P.A.3
  • 24
    • 84987850874 scopus 로고    scopus 로고
    • Sepsis pathophysiology, chronic critical illness, and persistent inflammation-immunosuppression and catabolism syndrome
    • Mira JC, Gentile LF, Mathias BJ, Sepsis pathophysiology, chronic critical illness, and persistent inflammation-immunosuppression and catabolism syndrome. Crit Care Med 2017; 45: 253–262
    • (2017) Crit Care Med , vol.45 , pp. 253-262
    • Mira, J.C.1    Gentile, L.F.2    Mathias, B.J.3
  • 25
    • 85056684786 scopus 로고    scopus 로고
    • Mortality in patients with septic shock by multidrug resistant bacteria
    • Epub ahead of print 18 January 2017
    • Busani S, Serafini G, Mantovani E, Mortality in patients with septic shock by multidrug resistant bacteria. J Intensive Care Med 2017. Epub ahead of print 18 January 2017. DOI: 10.1177/088506661668816
    • (2017) J Intensive Care Med
    • Busani, S.1    Serafini, G.2    Mantovani, E.3
  • 26
    • 0342626639 scopus 로고    scopus 로고
    • Impaired antigen presentation by human monocytes during endotoxin tolerance
    • Wolk K, Döcke WD, Baehr von V, Impaired antigen presentation by human monocytes during endotoxin tolerance. Blood 2000; 96: 218–223
    • (2000) Blood , vol.96 , pp. 218-223
    • Wolk, K.1    Döcke, W.D.2    Baehr von, V.3
  • 27
    • 2442501750 scopus 로고    scopus 로고
    • Monocyte human leukocyte antigen-DR transcriptional downregulation by cortisol during septic shock
    • Le Tulzo Y, Pangault C, Amiot L, Monocyte human leukocyte antigen-DR transcriptional downregulation by cortisol during septic shock. Am J Respir Crit Care Med 2004; 169: 1144–1151
    • (2004) Am J Respir Crit Care Med , vol.169 , pp. 1144-1151
    • Le Tulzo, Y.1    Pangault, C.2    Amiot, L.3
  • 28
    • 34250869345 scopus 로고    scopus 로고
    • Bench-to-bedside review: endotoxin tolerance as a model of leukocyte reprogramming in sepsis
    • Cavaillon JM, Adib-Conquy M. Bench-to-bedside review: endotoxin tolerance as a model of leukocyte reprogramming in sepsis. Crit Care 2006; 10: 233
    • (2006) Crit Care , vol.10 , pp. 233
    • Cavaillon, J.M.1    Adib-Conquy, M.2
  • 29
    • 33747156889 scopus 로고    scopus 로고
    • Persisting low monocyte human leukocyte antigen-DR expression predicts mortality in septic shock
    • Monneret G, Lepape A, Voirin N, Persisting low monocyte human leukocyte antigen-DR expression predicts mortality in septic shock. Intensive Care Med 2006; 32: 1175–1183
    • (2006) Intensive Care Med , vol.32 , pp. 1175-1183
    • Monneret, G.1    Lepape, A.2    Voirin, N.3
  • 30
    • 70349653291 scopus 로고    scopus 로고
    • Monocytic HLA-DR expression in intensive care patients: interest for prognosis and secondary infection prediction
    • Lukaszewicz A-C, Grienay M, Resche-Rigon M, Monocytic HLA-DR expression in intensive care patients: interest for prognosis and secondary infection prediction. Crit Care Med 2009; 37: 2746–2752
    • (2009) Crit Care Med , vol.37 , pp. 2746-2752
    • Lukaszewicz, A.-C.1    Grienay, M.2    Resche-Rigon, M.3
  • 31
    • 0041365362 scopus 로고    scopus 로고
    • Marked elevation of human circulating CD4+CD25+ regulatory T cells in sepsis-induced immunoparalysis
    • Monneret G, Debard A-L, Venet F, Marked elevation of human circulating CD4+CD25+ regulatory T cells in sepsis-induced immunoparalysis. Crit Care Med 2003; 31: 2068–2071
    • (2003) Crit Care Med , vol.31 , pp. 2068-2071
    • Monneret, G.1    Debard, A.-L.2    Venet, F.3
  • 32
    • 77954909395 scopus 로고    scopus 로고
    • Role of regulatory T cells in long-term immune dysfunction associated with severe sepsis
    • Nascimento DC, Alves-Filho JC, Sônego F, Role of regulatory T cells in long-term immune dysfunction associated with severe sepsis. Crit Care Med 2010; 38: 1718–1725
    • (2010) Crit Care Med , vol.38 , pp. 1718-1725
    • Nascimento, D.C.1    Alves-Filho, J.C.2    Sônego, F.3
  • 33
    • 34250359119 scopus 로고    scopus 로고
    • MyD88-dependent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polarization in sepsis
    • Delano MJ, Scumpia PO, Weinstein JS, MyD88-dependent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polarization in sepsis. J Exp Med 2007; 204: 1463–1474
    • (2007) J Exp Med , vol.204 , pp. 1463-1474
    • Delano, M.J.1    Scumpia, P.O.2    Weinstein, J.S.3
  • 34
    • 84891510610 scopus 로고    scopus 로고
    • Targeting the programmed cell death 1: programmed cell death ligand 1 pathway reverses T cell exhaustion in patients with sepsis
    • Chang K, Svabek C, Vazquez-Guillamet C, Targeting the programmed cell death 1: programmed cell death ligand 1 pathway reverses T cell exhaustion in patients with sepsis. Crit Care 2014; 18: R3
    • (2014) Crit Care , vol.18 , pp. R3
    • Chang, K.1    Svabek, C.2    Vazquez-Guillamet, C.3
  • 35
    • 16244393732 scopus 로고    scopus 로고
    • Sepsis-induced changes in macrophage co-stimulatory molecule expression: CD86 as a regulator of anti-inflammatory IL-10 response
    • Newton S, Ding Y, Chung C-S, Sepsis-induced changes in macrophage co-stimulatory molecule expression: CD86 as a regulator of anti-inflammatory IL-10 response. Surg Infect 2004; 5: 375–383
    • (2004) Surg Infect , vol.5 , pp. 375-383
    • Newton, S.1    Ding, Y.2    Chung, C.-S.3
  • 36
    • 67249123446 scopus 로고    scopus 로고
    • C5a mediates peripheral blood neutrophil dysfunction in critically ill patients
    • Conway Morris A, Kefala K, Wilkinson TS, C5a mediates peripheral blood neutrophil dysfunction in critically ill patients. Am J Respir Crit Care Med 2009; 180: 19–28
    • (2009) Am J Respir Crit Care Med , vol.180 , pp. 19-28
    • Conway Morris, A.1    Kefala, K.2    Wilkinson, T.S.3
  • 37
    • 1142310879 scopus 로고    scopus 로고
    • The dark side of C5a in sepsis
    • Ward PA. The dark side of C5a in sepsis. Nat Rev Immunol 2004; 4: 133–142
    • (2004) Nat Rev Immunol , vol.4 , pp. 133-142
    • Ward, P.A.1
  • 38
    • 29744447480 scopus 로고    scopus 로고
    • The effect of age on the development and outcome of adult sepsis
    • Martin GS, Mannino DM, Moss M. The effect of age on the development and outcome of adult sepsis. Crit Care Med 2006; 34: 15–21
    • (2006) Crit Care Med , vol.34 , pp. 15-21
    • Martin, G.S.1    Mannino, D.M.2    Moss, M.3
  • 39
    • 85009359407 scopus 로고    scopus 로고
    • Mechanisms underlying T Cell immunosenescence: aging and cytomegalovirus infection
    • Tu W, Rao S. Mechanisms underlying T Cell immunosenescence: aging and cytomegalovirus infection. Front Microbiol 2016; 7: 2111
    • (2016) Front Microbiol , vol.7 , pp. 2111
    • Tu, W.1    Rao, S.2
  • 40
    • 0029900294 scopus 로고    scopus 로고
    • Treatment of septic shock with the tumor necrosis factor receptor:Fc fusion protein. The Soluble TNF Receptor Sepsis Study Group
    • Fisher CJ, Agosti JM, Opal SM, Treatment of septic shock with the tumor necrosis factor receptor:Fc fusion protein. The Soluble TNF Receptor Sepsis Study Group. N Engl J Med 1996; 334: 1697–1702
    • (1996) N Engl J Med , vol.334 , pp. 1697-1702
    • Fisher, C.J.1    Agosti, J.M.2    Opal, S.M.3
  • 41
    • 27144509163 scopus 로고    scopus 로고
    • A network-based analysis of systemic inflammation in humans
    • Calvano SE, Xiao W, Richards DR, A network-based analysis of systemic inflammation in humans. Nature 2005; 437: 1032–1037
    • (2005) Nature , vol.437 , pp. 1032-1037
    • Calvano, S.E.1    Xiao, W.2    Richards, D.R.3
  • 42
    • 0034913364 scopus 로고    scopus 로고
    • Anti-tumor necrosis factor therapy in sepsis: update on clinical trials and lessons learned
    • Reinhart K, Karzai W. Anti-tumor necrosis factor therapy in sepsis: update on clinical trials and lessons learned. Crit Care Med 2001; 29: S121–S125
    • (2001) Crit Care Med , vol.29 , pp. S121-S125
    • Reinhart, K.1    Karzai, W.2
  • 43
    • 0036840629 scopus 로고    scopus 로고
    • Risk and the efficacy of antiinflammatory agents: retrospective and confirmatory studies of sepsis
    • Eichacker PQ, Parent C, Kalil A, Risk and the efficacy of antiinflammatory agents: retrospective and confirmatory studies of sepsis. Am J Respir Crit Care Med 2002; 166: 1197–1205
    • (2002) Am J Respir Crit Care Med , vol.166 , pp. 1197-1205
    • Eichacker, P.Q.1    Parent, C.2    Kalil, A.3
  • 44
  • 45
    • 84942583050 scopus 로고    scopus 로고
    • Procalcitonin versus C-reactive protein: Usefulness as biomarker of sepsis in ICU patient
    • Nargis W, Ibrahim M, Ahamed BU. Procalcitonin versus C-reactive protein: Usefulness as biomarker of sepsis in ICU patient. Int J Crit Illn Inj Sci 2014; 4: 195–199
    • (2014) Int J Crit Illn Inj Sci , vol.4 , pp. 195-199
    • Nargis, W.1    Ibrahim, M.2    Ahamed, B.U.3
  • 46
    • 41649099152 scopus 로고    scopus 로고
    • Guidelines for evaluation of new fever in critically ill adult patients: 2008 update from the American College of Critical Care Medicine and the Infectious Diseases Society of America
    • O'Grady NP, Barie PS, Bartlett JG, Guidelines for evaluation of new fever in critically ill adult patients: 2008 update from the American College of Critical Care Medicine and the Infectious Diseases Society of America. Crit Care Med 2008; 36: 1330–1349
    • (2008) Crit Care Med , vol.36 , pp. 1330-1349
    • O'Grady, N.P.1    Barie, P.S.2    Bartlett, J.G.3
  • 47
    • 84920593314 scopus 로고    scopus 로고
    • Prognostic evaluation of severe sepsis and septic shock: procalcitonin clearance vs Δ Sequential Organ Failure Assessment
    • de Azevedo JRA, Torres OJM, Beraldi RA, Prognostic evaluation of severe sepsis and septic shock: procalcitonin clearance vs Δ Sequential Organ Failure Assessment. J Crit Care 2015; 30: 219.e9–219.e12
    • (2015) J Crit Care , vol.30
    • de Azevedo, J.R.A.1    Torres, O.J.M.2    Beraldi, R.A.3
  • 48
    • 84892964775 scopus 로고    scopus 로고
    • The efficacy of procalcitonin as a biomarker in the management of sepsis: slaying dragons or tilting at windmills?
    • Sridharan P, Chamberlain RS. The efficacy of procalcitonin as a biomarker in the management of sepsis: slaying dragons or tilting at windmills? Surg Infect 2013; 14: 489–511
    • (2013) Surg Infect , vol.14 , pp. 489-511
    • Sridharan, P.1    Chamberlain, R.S.2
  • 49
    • 0028129179 scopus 로고
    • Interleukin 6 is a prognostic indicator of outcome in severe intra-abdominal sepsis
    • Patel RT, Deen KI, Youngs D, Interleukin 6 is a prognostic indicator of outcome in severe intra-abdominal sepsis. Br J Surg 1994; 81: 1306–1308
    • (1994) Br J Surg , vol.81 , pp. 1306-1308
    • Patel, R.T.1    Deen, K.I.2    Youngs, D.3
  • 50
    • 0033969341 scopus 로고    scopus 로고
    • Pro- versus anti-inflammatory cytokine profile in patients with severe sepsis: a marker for prognosis and future therapeutic options
    • Gogos CA, Drosou E, Bassaris HP, Pro- versus anti-inflammatory cytokine profile in patients with severe sepsis: a marker for prognosis and future therapeutic options. J Infect Dis 2000; 181: 176–180
    • (2000) J Infect Dis , vol.181 , pp. 176-180
    • Gogos, C.A.1    Drosou, E.2    Bassaris, H.P.3
  • 51
    • 84871881145 scopus 로고    scopus 로고
    • Serial cytokine levels in patients with severe sepsis
    • Wu H-P, Chen C-K, Chung K, Serial cytokine levels in patients with severe sepsis. Inflamm Res 2009; 58: 385–393
    • (2009) Inflamm Res , vol.58 , pp. 385-393
    • Wu, H.-P.1    Chen, C.-K.2    Chung, K.3
  • 52
    • 34249318067 scopus 로고    scopus 로고
    • Cytokine profiles as markers of disease severity in sepsis: a multiplex analysis
    • Bozza FA, Salluh JI, Japiassu AM, Cytokine profiles as markers of disease severity in sepsis: a multiplex analysis. Crit Care 2007; 11: R49
    • (2007) Crit Care , vol.11 , pp. R49
    • Bozza, F.A.1    Salluh, J.I.2    Japiassu, A.M.3
  • 53
    • 33745475888 scopus 로고    scopus 로고
    • Development and validation of a multiplex add-on assay for sepsis biomarkers using xMAP technology
    • Kofoed K, Schneider UV, Scheel T, Development and validation of a multiplex add-on assay for sepsis biomarkers using xMAP technology. Clin Chem 2006; 52: 1284–1293
    • (2006) Clin Chem , vol.52 , pp. 1284-1293
    • Kofoed, K.1    Schneider, U.V.2    Scheel, T.3
  • 54
    • 59649123701 scopus 로고    scopus 로고
    • A prospective, multicenter derivation of a biomarker panel to assess risk of organ dysfunction, shock, and death in emergency department patients with suspected sepsis
    • Shapiro NI, Trzeciak S, Hollander JE, A prospective, multicenter derivation of a biomarker panel to assess risk of organ dysfunction, shock, and death in emergency department patients with suspected sepsis. Crit Care Med 2009; 37: 96–104
    • (2009) Crit Care Med , vol.37 , pp. 96-104
    • Shapiro, N.I.1    Trzeciak, S.2    Hollander, J.E.3
  • 55
    • 84963686144 scopus 로고    scopus 로고
    • Plasma cytokine levels predict response to corticosteroids in septic shock
    • Bentzer P, Fjell C, Walley KR, Plasma cytokine levels predict response to corticosteroids in septic shock. Intensive Care Med 2016; 42: 1970–1979
    • (2016) Intensive Care Med , vol.42 , pp. 1970-1979
    • Bentzer, P.1    Fjell, C.2    Walley, K.R.3
  • 56
    • 0027451272 scopus 로고
    • Lipopolysaccharide structure-function relationship in activation versus reprogramming of mouse peritoneal macrophages
    • Zhang X, Morrison DC. Lipopolysaccharide structure-function relationship in activation versus reprogramming of mouse peritoneal macrophages. J Leukoc Biol 1993; 54: 444–450
    • (1993) J Leukoc Biol , vol.54 , pp. 444-450
    • Zhang, X.1    Morrison, D.C.2
  • 57
    • 84868300033 scopus 로고    scopus 로고
    • Reversal of immunoparalysis in humans in vivo: a double-blind, placebo-controlled, randomized pilot study
    • Leentjens J, Kox M, Koch RM, Reversal of immunoparalysis in humans in vivo: a double-blind, placebo-controlled, randomized pilot study. Am J Respir Crit Care Med 2012; 186: 838–845
    • (2012) Am J Respir Crit Care Med , vol.186 , pp. 838-845
    • Leentjens, J.1    Kox, M.2    Koch, R.M.3
  • 58
    • 84964225061 scopus 로고    scopus 로고
    • Stimulated whole blood cytokine release as a biomarker of immunosuppression in the critically ill: the need for a standardized methodology
    • Segre E, Fullerton JN. Stimulated whole blood cytokine release as a biomarker of immunosuppression in the critically ill: the need for a standardized methodology. Shock 2016; 45: 490–494
    • (2016) Shock , vol.45 , pp. 490-494
    • Segre, E.1    Fullerton, J.N.2
  • 59
    • 3042818432 scopus 로고    scopus 로고
    • Plasma level of a triggering receptor expressed on myeloid cells-1: its diagnostic accuracy in patients with suspected sepsis
    • Gibot S, Kolopp-Sarda M-N, Béné MC, Plasma level of a triggering receptor expressed on myeloid cells-1: its diagnostic accuracy in patients with suspected sepsis. Ann Intern Med 2004; 141: 9–15
    • (2004) Ann Intern Med , vol.141 , pp. 9-15
    • Gibot, S.1    Kolopp-Sarda, M.-N.2    Béné, M.C.3
  • 60
    • 84862827407 scopus 로고    scopus 로고
    • Measurement of plasma sTREM-1 in patients with severe sepsis receiving early goal-directed therapy and evaluation of its usefulness
    • Jeong SJ, Song YG, Kim CO, Measurement of plasma sTREM-1 in patients with severe sepsis receiving early goal-directed therapy and evaluation of its usefulness. Shock 2012; 37: 574–578
    • (2012) Shock , vol.37 , pp. 574-578
    • Jeong, S.J.1    Song, Y.G.2    Kim, C.O.3
  • 61
    • 84971280532 scopus 로고    scopus 로고
    • Role of sTREM-1 in predicting mortality of infection: a systematic review and meta-analysis
    • Su L, Liu D, Chai W, Role of sTREM-1 in predicting mortality of infection: a systematic review and meta-analysis. BMJ Open 2016; 6: e010314
    • (2016) BMJ Open , vol.6 , pp. e010314
    • Su, L.1    Liu, D.2    Chai, W.3
  • 62
    • 0030917203 scopus 로고    scopus 로고
    • Monocyte deactivation in septic patients: restoration by IFN-γ treatment
    • Döcke WD, Randow F, Syrbe U, Monocyte deactivation in septic patients: restoration by IFN-γ treatment. Nat Med 1997; 3: 678–681
    • (1997) Nat Med , vol.3 , pp. 678-681
    • Döcke, W.D.1    Randow, F.2    Syrbe, U.3
  • 63
    • 0036310152 scopus 로고    scopus 로고
    • Immunoparalysis in patients with severe trauma and the effect of inhaled interferon-γ
    • Nakos G, Malamou-Mitsi VD, Lachana A, Immunoparalysis in patients with severe trauma and the effect of inhaled interferon-γ. Crit Care Med 2002; 30: 1488–1494
    • (2002) Crit Care Med , vol.30 , pp. 1488-1494
    • Nakos, G.1    Malamou-Mitsi, V.D.2    Lachana, A.3
  • 64
    • 84879314541 scopus 로고    scopus 로고
    • Inter-laboratory assessment of flow cytometric monocyte HLA-DR expression in clinical samples
    • Demaret J, Walencik A, Jacob M-C, Inter-laboratory assessment of flow cytometric monocyte HLA-DR expression in clinical samples. Cytometry B Clin Cytom 2013; 84: 59–62
    • (2013) Cytometry B Clin Cytom , vol.84 , pp. 59-62
    • Demaret, J.1    Walencik, A.2    Jacob, M.-C.3
  • 65
    • 70349469854 scopus 로고    scopus 로고
    • Granulocyte-macrophage colony-stimulating factor to reverse sepsis-associated immunosuppression: a double-blind, randomized, placebo-controlled multicenter trial
    • Meisel C, Schefold JC, Pschowski R, Granulocyte-macrophage colony-stimulating factor to reverse sepsis-associated immunosuppression: a double-blind, randomized, placebo-controlled multicenter trial. Am J Respir Crit Care Med 2009; 180: 640–648
    • (2009) Am J Respir Crit Care Med , vol.180 , pp. 640-648
    • Meisel, C.1    Schefold, J.C.2    Pschowski, R.3
  • 66
    • 84992220369 scopus 로고    scopus 로고
    • Comparison of monocyte human leukocyte antigen-DR expression and stimulated tumor necrosis factor alpha production as outcome predictors in severe sepsis: a prospective observational study
    • Drewry AM, Ablordeppey EA, Murray ET, Comparison of monocyte human leukocyte antigen-DR expression and stimulated tumor necrosis factor alpha production as outcome predictors in severe sepsis: a prospective observational study. Crit Care 2016; 20: 334
    • (2016) Crit Care , vol.20 , pp. 334
    • Drewry, A.M.1    Ablordeppey, E.A.2    Murray, E.T.3
  • 67
    • 77957690699 scopus 로고    scopus 로고
    • Early assessment of leukocyte alterations at diagnosis of septic shock
    • Venet F, Davin F, Guignant C, Early assessment of leukocyte alterations at diagnosis of septic shock. Shock 2010; 34: 358–363
    • (2010) Shock , vol.34 , pp. 358-363
    • Venet, F.1    Davin, F.2    Guignant, C.3
  • 68
    • 84930084195 scopus 로고    scopus 로고
    • Persistent lymphopenia after diagnosis of sepsis predicts mortality
    • Drewry AM, Samra N, Skrupky LP, Persistent lymphopenia after diagnosis of sepsis predicts mortality. Shock 2014; 42: 383–391
    • (2014) Shock , vol.42 , pp. 383-391
    • Drewry, A.M.1    Samra, N.2    Skrupky, L.P.3
  • 69
    • 84927150740 scopus 로고    scopus 로고
    • Immune checkpoint blockade in cancer therapy
    • Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol 2015; 33: 1974–1982
    • (2015) J Clin Oncol , vol.33 , pp. 1974-1982
    • Postow, M.A.1    Callahan, M.K.2    Wolchok, J.D.3
  • 70
    • 84858766182 scopus 로고    scopus 로고
    • The blockade of immune checkpoints in cancer immunotherapy
    • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252–264
    • (2012) Nat Rev Cancer , vol.12 , pp. 252-264
    • Pardoll, D.M.1
  • 71
    • 84973375128 scopus 로고    scopus 로고
    • Monocyte programmed death ligand-1 expression after 3-4 days of sepsis is associated with risk stratification and mortality in septic patients: a prospective cohort study
    • Shao R, Fang Y, Yu H, Monocyte programmed death ligand-1 expression after 3-4 days of sepsis is associated with risk stratification and mortality in septic patients: a prospective cohort study. Crit Care 2016; 20: 124
    • (2016) Crit Care , vol.20 , pp. 124
    • Shao, R.1    Fang, Y.2    Yu, H.3
  • 72
    • 79953121419 scopus 로고    scopus 로고
    • Programmed death-1 levels correlate with increased mortality, nosocomial infection and immune dysfunctions in septic shock patients
    • Guignant C, Lepape A, Huang X, Programmed death-1 levels correlate with increased mortality, nosocomial infection and immune dysfunctions in septic shock patients. Crit Care 2011; 15: R99
    • (2011) Crit Care , vol.15 , pp. R99
    • Guignant, C.1    Lepape, A.2    Huang, X.3
  • 73
    • 84862766869 scopus 로고    scopus 로고
    • A prospective analysis of lymphocyte phenotype and function over the course of acute sepsis
    • Boomer JS, Shuherk-Shaffer J, Hotchkiss RS, A prospective analysis of lymphocyte phenotype and function over the course of acute sepsis. Crit Care 2012; 16: R112
    • (2012) Crit Care , vol.16 , pp. R112
    • Boomer, J.S.1    Shuherk-Shaffer, J.2    Hotchkiss, R.S.3
  • 74
    • 84924952897 scopus 로고    scopus 로고
    • Human monocytes undergo functional re-programming during sepsis mediated by hypoxia-inducible factor-1α
    • Shalova IN, Lim JY, Chittezhath M, Human monocytes undergo functional re-programming during sepsis mediated by hypoxia-inducible factor-1α. Immunity 2015; 42: 484–498
    • (2015) Immunity , vol.42 , pp. 484-498
    • Shalova, I.N.1    Lim, J.Y.2    Chittezhath, M.3
  • 75
    • 85056688868 scopus 로고    scopus 로고
    • Association between mRNA expression of CD74 and IL10 and risk of ICU-acquired infections: a multicenter cohort study
    • Peronnet E, Venet F, Maucort-Boulch D, Association between mRNA expression of CD74 and IL10 and risk of ICU-acquired infections: a multicenter cohort study. Intensive Care Med 2017; 111: 778–778
    • (2017) Intensive Care Med , vol.111 , pp. 778
    • Peronnet, E.1    Venet, F.2    Maucort-Boulch, D.3
  • 76
    • 84891639053 scopus 로고    scopus 로고
    • Decreased HLA-DR antigen-associated invariant chain (CD74) mRNA expression predicts mortality after septic shock
    • Cazalis M-A, Friggeri A, Cavé L, Decreased HLA-DR antigen-associated invariant chain (CD74) mRNA expression predicts mortality after septic shock. Crit Care 2013; 17: R287
    • (2013) Crit Care , vol.17 , pp. R287
    • Cazalis, M.-A.1    Friggeri, A.2    Cavé, L.3
  • 77
    • 84953790265 scopus 로고    scopus 로고
    • A molecular host response assay to discriminate between sepsis and infection-negative systemic inflammation in critically ill patients: discovery and validation in independent cohorts
    • McHugh L, Seldon TA, Brandon RA, A molecular host response assay to discriminate between sepsis and infection-negative systemic inflammation in critically ill patients: discovery and validation in independent cohorts. PLoS Med 2015; 12: e1001916
    • (2015) PLoS Med , vol.12 , pp. e1001916
    • McHugh, L.1    Seldon, T.A.2    Brandon, R.A.3
  • 78
    • 85026823884 scopus 로고    scopus 로고
    • Shared and distinct aspects of the sepsis transcriptomic response to fecal peritonitis and pneumonia
    • Burnham KL, Davenport EE, Radhakrishnan J, Shared and distinct aspects of the sepsis transcriptomic response to fecal peritonitis and pneumonia. Am J Respir Crit Care Med 2017; 196: 328–339
    • (2017) Am J Respir Crit Care Med , vol.196 , pp. 328-339
    • Burnham, K.L.1    Davenport, E.E.2    Radhakrishnan, J.3
  • 79
    • 84979599328 scopus 로고    scopus 로고
    • Predictive value of cell-surface markers in infections in critically ill patients: protocol for an observational study (ImmuNe FailurE in Critical Therapy (INFECT) Study)
    • Conway Morris A, Datta D, Shankar-Hari M, Predictive value of cell-surface markers in infections in critically ill patients: protocol for an observational study (ImmuNe FailurE in Critical Therapy (INFECT) Study). BMJ Open 2016; 6: e011326
    • (2016) BMJ Open , vol.6 , pp. e011326
    • Conway Morris, A.1    Datta, D.2    Shankar-Hari, M.3
  • 80
    • 84891637868 scopus 로고    scopus 로고
    • Monocyte HLA-DR in sepsis: shall we stop following the flow?
    • Monneret G, Venet F. Monocyte HLA-DR in sepsis: shall we stop following the flow? Crit Care 2014; 18: 102
    • (2014) Crit Care , vol.18 , pp. 102
    • Monneret, G.1    Venet, F.2
  • 81
    • 84884967732 scopus 로고    scopus 로고
    • Preliminary results in quantitation of HLA-DRA by real-time PCR: a promising approach to identify immunosuppression in sepsis
    • Cajander S, Bäckman A, Tina E, Preliminary results in quantitation of HLA-DRA by real-time PCR: a promising approach to identify immunosuppression in sepsis. Crit Care 2013; 17: R223
    • (2013) Crit Care , vol.17 , pp. R223
    • Cajander, S.1    Bäckman, A.2    Tina, E.3
  • 83
    • 85040932622 scopus 로고    scopus 로고
    • Comparison of cd64 levels performed by the FACS and Accellix systems
    • Sprung CL, Morales RC, Kasdan H, Comparison of cd64 levels performed by the FACS and Accellix systems. Intensive Care Med Exp 2015; 3: A1012
    • (2015) Intensive Care Med Exp , vol.3 , pp. A1012
    • Sprung, C.L.1    Morales, R.C.2    Kasdan, H.3
  • 84
    • 84862859820 scopus 로고    scopus 로고
    • Safety, activity, and immune correlates of anti-PD-1 antibody in cancer
    • Topalian SL, Hodi FS, Brahmer JR, Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366: 2443–2454
    • (2012) N Engl J Med , vol.366 , pp. 2443-2454
    • Topalian, S.L.1    Hodi, F.S.2    Brahmer, J.R.3
  • 85
    • 84904549329 scopus 로고    scopus 로고
    • Parallels between cancer and infectious disease
    • Hotchkiss RS, Moldawer LL. Parallels between cancer and infectious disease. N Engl J Med 2014; 371: 380–383
    • (2014) N Engl J Med , vol.371 , pp. 380-383
    • Hotchkiss, R.S.1    Moldawer, L.L.2
  • 86
    • 85008154354 scopus 로고    scopus 로고
    • Nivolumab plus interferon-γ in the treatment of intractable mucormycosis
    • Grimaldi D, Pradier O, Hotchkiss RS, Nivolumab plus interferon-γ in the treatment of intractable mucormycosis. Lancet Infect Dis 2017; 17: 18
    • (2017) Lancet Infect Dis , vol.17 , pp. 18
    • Grimaldi, D.1    Pradier, O.2    Hotchkiss, R.S.3
  • 87
    • 85017417748 scopus 로고    scopus 로고
    • Adjuvant interferon-gamma immunotherapy in a patient with progressive cerebral Nocardia abscesses
    • Leentjens J, Gresnigt MS, van de Veerdonk FL, Adjuvant interferon-gamma immunotherapy in a patient with progressive cerebral Nocardia abscesses. Int J Infect Dis 2017; 59: 25–28
    • (2017) Int J Infect Dis , vol.59 , pp. 25-28
    • Leentjens, J.1    Gresnigt, M.S.2    van de Veerdonk, F.L.3
  • 88
    • 84924976132 scopus 로고    scopus 로고
    • Interleukin 7 and anti-programmed cell death 1 antibody have differing effects to reverse sepsis-induced immunosuppression
    • Shindo Y, Unsinger J, Burnham C-A, Interleukin 7 and anti-programmed cell death 1 antibody have differing effects to reverse sepsis-induced immunosuppression. Shock 2015; 43: 334–343
    • (2015) Shock , vol.43 , pp. 334-343
    • Shindo, Y.1    Unsinger, J.2    Burnham, C.-A.3
  • 89
    • 84877262213 scopus 로고    scopus 로고
    • Blockade of the negative co-stimulatory molecules PD-1 and CTLA-4 improves survival in primary and secondary fungal sepsis
    • Chang KC, Burnham C-A, Compton SM, Blockade of the negative co-stimulatory molecules PD-1 and CTLA-4 improves survival in primary and secondary fungal sepsis. Crit Care 2013; 17: R85
    • (2013) Crit Care , vol.17 , pp. R85
    • Chang, K.C.1    Burnham, C.-A.2    Compton, S.M.3
  • 90
    • 32544459770 scopus 로고    scopus 로고
    • T cells during chronic viral infection
    • Barber DL, Wherry EJ, Masopust D, T cells during chronic viral infection. Nature 2006; 439: 682–687
    • (2006) Nature , vol.439 , pp. 682-687
    • Barber, D.L.1    Wherry, E.J.2    Masopust, D.3
  • 91
    • 84878089612 scopus 로고    scopus 로고
    • A randomized, double-blind, placebo-controlled assessment of BMS-936558, a fully human monoclonal antibody to programmed death-1 (PD-1), in patients with chronic hepatitis C virus infection
    • Gardiner D, Lalezari J, Lawitz E, A randomized, double-blind, placebo-controlled assessment of BMS-936558, a fully human monoclonal antibody to programmed death-1 (PD-1), in patients with chronic hepatitis C virus infection. PLoS One 2013; 8: e63818
    • (2013) PLoS One , vol.8 , pp. e63818
    • Gardiner, D.1    Lalezari, J.2    Lawitz, E.3
  • 92
    • 84883746616 scopus 로고    scopus 로고
    • Immunotherapy of chronic hepatitis C virus infection with antibodies against programmed cell death-1 (PD-1)
    • Fuller MJ, Callendret B, Zhu B, Immunotherapy of chronic hepatitis C virus infection with antibodies against programmed cell death-1 (PD-1). Proc Natl Acad Sci USA 2013; 110: 15001–15006
    • (2013) Proc Natl Acad Sci USA , vol.110 , pp. 15001-15006
    • Fuller, M.J.1    Callendret, B.2    Zhu, B.3
  • 93
    • 85019137781 scopus 로고    scopus 로고
    • A current perspective on cancer immune therapy: step-by-step approach to constructing the magic bullet
    • D'Errico G, Machado HL, Sainz B. A current perspective on cancer immune therapy: step-by-step approach to constructing the magic bullet. Clin Transl Med 2017; 6: 3
    • (2017) Clin Transl Med , vol.6 , pp. 3
    • D'Errico, G.1    Machado, H.L.2    Sainz, B.3
  • 94
    • 79959716811 scopus 로고    scopus 로고
    • Dose-dependent effect of anti-CTLA-4 on survival in sepsis
    • Inoue S, Bo L, Bian J, Dose-dependent effect of anti-CTLA-4 on survival in sepsis. Shock 2011; 36: 38–44
    • (2011) Shock , vol.36 , pp. 38-44
    • Inoue, S.1    Bo, L.2    Bian, J.3
  • 95
    • 78650614116 scopus 로고    scopus 로고
    • IFN-γ abrogates endotoxin tolerance by facilitating Toll-like receptor-induced chromatin remodeling
    • Chen J, Ivashkiv LB. IFN-γ abrogates endotoxin tolerance by facilitating Toll-like receptor-induced chromatin remodeling. Proc Natl Acad Sci USA 2010; 107: 19438–19443
    • (2010) Proc Natl Acad Sci USA , vol.107 , pp. 19438-19443
    • Chen, J.1    Ivashkiv, L.B.2
  • 96
    • 77955274188 scopus 로고    scopus 로고
    • IFN-γ enhances killing of methicillin-resistant Staphylococcus aureus by human monocytes more effectively than GM-CSF in the presence of daptomycin and other antibiotics
    • Smith RP, Baltch AL, Ritz WJ, IFN-γ enhances killing of methicillin-resistant Staphylococcus aureus by human monocytes more effectively than GM-CSF in the presence of daptomycin and other antibiotics. Cytokine 2010; 51: 274–277
    • (2010) Cytokine , vol.51 , pp. 274-277
    • Smith, R.P.1    Baltch, A.L.2    Ritz, W.J.3
  • 97
    • 0031569105 scopus 로고    scopus 로고
    • In vitro prevention and reversal of lipopolysaccharide desensitization by IFN-gamma, IL-12, and granulocyte-macrophage colony-stimulating factor
    • Randow F, Döcke WD, Bundschuh DS, In vitro prevention and reversal of lipopolysaccharide desensitization by IFN-gamma, IL-12, and granulocyte-macrophage colony-stimulating factor. J Immunol 1997; 158: 2911–2918
    • (1997) J Immunol , vol.158 , pp. 2911-2918
    • Randow, F.1    Döcke, W.D.2    Bundschuh, D.S.3
  • 98
    • 0031054684 scopus 로고    scopus 로고
    • Interferon Gamma-1b in the treatment of compensatory anti-inflammatory response syndrome
    • Kox WJ. Interferon Gamma-1b in the treatment of compensatory anti-inflammatory response syndrome. Arch Intern Med 1997; 157: 389
    • (1997) Arch Intern Med , vol.157 , pp. 389
    • Kox, W.J.1
  • 99
    • 0023623568 scopus 로고
    • Anti-interferon-gamma antibody protects mice against the generalized Shwartzman reaction
    • Billiau A, Heremans H, Vandekerckhove F, Anti-interferon-gamma antibody protects mice against the generalized Shwartzman reaction. Eur J Immunol 1987; 17: 1851–1854
    • (1987) Eur J Immunol , vol.17 , pp. 1851-1854
    • Billiau, A.1    Heremans, H.2    Vandekerckhove, F.3
  • 100
    • 80054852191 scopus 로고    scopus 로고
    • mRNA-based approach to monitor recombinant gamma-interferon restoration of LPS-induced endotoxin tolerance
    • Turrel-Davin F, Venet F, Monnin C, mRNA-based approach to monitor recombinant gamma-interferon restoration of LPS-induced endotoxin tolerance. Crit Care 2011; 15: R252
    • (2011) Crit Care , vol.15 , pp. R252
    • Turrel-Davin, F.1    Venet, F.2    Monnin, C.3
  • 102
    • 84861529161 scopus 로고    scopus 로고
    • Adjunctive interferon-γ immunotherapy for the treatment of HIV-associated cryptococcal meningitis: a randomized controlled trial
    • Jarvis JN, Meintjes G, Rebe K, Adjunctive interferon-γ immunotherapy for the treatment of HIV-associated cryptococcal meningitis: a randomized controlled trial. AIDS 2012; 26: 1105–1113
    • (2012) AIDS , vol.26 , pp. 1105-1113
    • Jarvis, J.N.1    Meintjes, G.2    Rebe, K.3
  • 103
    • 84897530707 scopus 로고    scopus 로고
    • Interferon-gamma as adjunctive immunotherapy for invasive fungal infections: a case series
    • Delsing CE, Gresnigt MS, Leentjens J, Interferon-gamma as adjunctive immunotherapy for invasive fungal infections: a case series. BMC Infect Dis 2014; 14: 166
    • (2014) BMC Infect Dis , vol.14 , pp. 166
    • Delsing, C.E.1    Gresnigt, M.S.2    Leentjens, J.3
  • 104
    • 84874256945 scopus 로고    scopus 로고
    • Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach
    • Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis 2013; 13: 260–268
    • (2013) Lancet Infect Dis , vol.13 , pp. 260-268
    • Hotchkiss, R.S.1    Monneret, G.2    Payen, D.3
  • 105
    • 84859837162 scopus 로고    scopus 로고
    • Regulation of dendritic cell development by GM-CSF: molecular control and implications for immune homeostasis and therapy
    • van de Laar L, Coffer PJ, Woltman AM. Regulation of dendritic cell development by GM-CSF: molecular control and implications for immune homeostasis and therapy. Blood 2012; 119: 3383–3393
    • (2012) Blood , vol.119 , pp. 3383-3393
    • van de Laar, L.1    Coffer, P.J.2    Woltman, A.M.3
  • 106
    • 0029023295 scopus 로고
    • GM-CSF rapidly primes mice for enhanced cytokine production in response to LPS and TNF
    • Brissette WH, Baker DA, Stam EJ, GM-CSF rapidly primes mice for enhanced cytokine production in response to LPS and TNF. Cytokine 1995; 7: 291–295
    • (1995) Cytokine , vol.7 , pp. 291-295
    • Brissette, W.H.1    Baker, D.A.2    Stam, E.J.3
  • 107
    • 0033609093 scopus 로고    scopus 로고
    • Granulocyte-macrophage colony stimulating factor prevents the progression of atherosclerosis via changes in the cellular and extracellular composition of atherosclerotic lesions in Watanabe heritable hyperlipidemic rabbits
    • Shindo J, Ishibashi T, Yokoyama K, Granulocyte-macrophage colony stimulating factor prevents the progression of atherosclerosis via changes in the cellular and extracellular composition of atherosclerotic lesions in Watanabe heritable hyperlipidemic rabbits. Circulation 1999; 99: 2150–2156
    • (1999) Circulation , vol.99 , pp. 2150-2156
    • Shindo, J.1    Ishibashi, T.2    Yokoyama, K.3
  • 108
    • 0027444828 scopus 로고    scopus 로고
    • Role of granulocyte-macrophage colony-stimulating factor in pulmonary fibrosis induced in mice by bleomycin
    • Piguet PF, Grau GE, de Kossodo S. Role of granulocyte-macrophage colony-stimulating factor in pulmonary fibrosis induced in mice by bleomycin. Experimental Lung Res 2009; 19: 579–587
    • (2009) Experimental Lung Res , vol.19 , pp. 579-587
    • Piguet, P.F.1    Grau, G.E.2    de Kossodo, S.3
  • 109
    • 4344590947 scopus 로고    scopus 로고
    • High-dose granulocyte-macrophage colony-stimulating factor-producing vaccines impair the immune response through the recruitment of myeloid suppressor cells
    • Serafini P, Carbley R, Noonan KA, High-dose granulocyte-macrophage colony-stimulating factor-producing vaccines impair the immune response through the recruitment of myeloid suppressor cells. Cancer Res 2004; 64: 6337–6343
    • (2004) Cancer Res , vol.64 , pp. 6337-6343
    • Serafini, P.1    Carbley, R.2    Noonan, K.A.3
  • 110
    • 79955418828 scopus 로고    scopus 로고
    • Harnessing the biology of IL-7 for therapeutic application
    • Mackall CL, Fry TJ, Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol 2011; 11: 330–342
    • (2011) Nat Rev Immunol , vol.11 , pp. 330-342
    • Mackall, C.L.1    Fry, T.J.2    Gress, R.E.3
  • 111
    • 84861190857 scopus 로고    scopus 로고
    • The many roles of IL-7 in B cell development; mediator of survival, proliferation and differentiation
    • Corfe SA, Paige CJ. The many roles of IL-7 in B cell development; mediator of survival, proliferation and differentiation. Semin Immunol 2012; 24: 198–208
    • (2012) Semin Immunol , vol.24 , pp. 198-208
    • Corfe, S.A.1    Paige, C.J.2
  • 112
    • 84861194822 scopus 로고    scopus 로고
    • Interleukin 7, maestro of the immune system
    • Sprent J, Surh CD. Interleukin 7, maestro of the immune system. Semin Immunol 2012; 24: 149–150
    • (2012) Semin Immunol , vol.24 , pp. 149-150
    • Sprent, J.1    Surh, C.D.2
  • 113
    • 65249105408 scopus 로고    scopus 로고
    • Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment
    • Levy Y, Lacabaratz C, Weiss L, Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment. J Clin Invest 2009; 119: 997–1007
    • (2009) J Clin Invest , vol.119 , pp. 997-1007
    • Levy, Y.1    Lacabaratz, C.2    Weiss, L.3
  • 114
    • 67349193717 scopus 로고    scopus 로고
    • Adjuvant IL-7 antagonizes multiple cellular and molecular inhibitory networks to enhance immunotherapies
    • Pellegrini M, Calzascia T, Elford AR, Adjuvant IL-7 antagonizes multiple cellular and molecular inhibitory networks to enhance immunotherapies. Nat Med 2009; 15: 528–536
    • (2009) Nat Med , vol.15 , pp. 528-536
    • Pellegrini, M.1    Calzascia, T.2    Elford, A.R.3
  • 115
    • 84860771152 scopus 로고    scopus 로고
    • Interleukin-7 and immune reconstitution in cancer patients: a new paradigm for dramatically increasing overall survival
    • Morre M, Beq S. Interleukin-7 and immune reconstitution in cancer patients: a new paradigm for dramatically increasing overall survival. Target Oncol 2012; 7: 55–68
    • (2012) Target Oncol , vol.7 , pp. 55-68
    • Morre, M.1    Beq, S.2
  • 116
    • 84864125153 scopus 로고    scopus 로고
    • Effects of recombinant human interleukin 7 on T-cell recovery and thymic output in HIV-infected patients receiving antiretroviral therapy: results of a phase I/IIa randomized, placebo-controlled, multicenter study
    • Lévy Y, Sereti I, Tambussi G, Effects of recombinant human interleukin 7 on T-cell recovery and thymic output in HIV-infected patients receiving antiretroviral therapy: results of a phase I/IIa randomized, placebo-controlled, multicenter study. Clin Infect Dis 2012; 55: 291–300
    • (2012) Clin Infect Dis , vol.55 , pp. 291-300
    • Lévy, Y.1    Sereti, I.2    Tambussi, G.3
  • 117
    • 46949105036 scopus 로고    scopus 로고
    • Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets
    • Sportès C, Hakim FT, Memon SA, Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. J Exp Med 2008; 205: 1701–1714
    • (2008) J Exp Med , vol.205 , pp. 1701-1714
    • Sportès, C.1    Hakim, F.T.2    Memon, S.A.3
  • 118
    • 84866936380 scopus 로고    scopus 로고
    • Interleukin-7 ameliorates immune dysfunction and improves survival in a 2-hit model of fungal sepsis
    • Unsinger J, Burnham C-AD, McDonough J, Interleukin-7 ameliorates immune dysfunction and improves survival in a 2-hit model of fungal sepsis. J Infect Dis 2012; 206: 606–616
    • (2012) J Infect Dis , vol.206 , pp. 606-616
    • Unsinger, J.1    Burnham, C.-A.D.2    McDonough, J.3
  • 119
    • 85011320845 scopus 로고    scopus 로고
    • Interleukin 7 immunotherapy improves host immunity and survival in a two-hit model of Pseudomonas aeruginosa pneumonia
    • Shindo Y, Fuchs AG, Davis CG, Interleukin 7 immunotherapy improves host immunity and survival in a two-hit model of Pseudomonas aeruginosa pneumonia. J Leukoc Biol 2017; 101: 543–554
    • (2017) J Leukoc Biol , vol.101 , pp. 543-554
    • Shindo, Y.1    Fuchs, A.G.2    Davis, C.G.3
  • 120
    • 77951648850 scopus 로고    scopus 로고
    • IL-7 promotes T cell viability, trafficking, and functionality and improves survival in sepsis
    • Unsinger J, McGlynn M, Kasten KR, IL-7 promotes T cell viability, trafficking, and functionality and improves survival in sepsis. J Immunol 2010; 184: 3768–3779
    • (2010) J Immunol , vol.184 , pp. 3768-3779
    • Unsinger, J.1    McGlynn, M.2    Kasten, K.R.3
  • 121
    • 78049433417 scopus 로고    scopus 로고
    • Interleukin-7 (IL-7) treatment accelerates neutrophil recruitment through gamma delta T-cell IL-17 production in a murine model of sepsis
    • Kasten KR, Prakash PS, Unsinger J, Interleukin-7 (IL-7) treatment accelerates neutrophil recruitment through gamma delta T-cell IL-17 production in a murine model of sepsis. Infect Immun 2010; 78: 4714–4722
    • (2010) Infect Immun , vol.78 , pp. 4714-4722
    • Kasten, K.R.1    Prakash, P.S.2    Unsinger, J.3
  • 122
    • 18944379196 scopus 로고    scopus 로고
    • The many faces of IL-7: from lymphopoiesis to peripheral T cell maintenance
    • Fry TJ, Mackall CL. The many faces of IL-7: from lymphopoiesis to peripheral T cell maintenance. J Immunol 2005; 174: 6571–6576
    • (2005) J Immunol , vol.174 , pp. 6571-6576
    • Fry, T.J.1    Mackall, C.L.2
  • 123
    • 65249105408 scopus 로고    scopus 로고
    • Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment
    • Levy Y, Lacabaratz C, Weiss L, Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment. J Clin Invest 2009; 119: 997–1007
    • (2009) J Clin Invest , vol.119 , pp. 997-1007
    • Levy, Y.1    Lacabaratz, C.2    Weiss, L.3
  • 124
    • 67650590873 scopus 로고    scopus 로고
    • IL-7 administration drives T cell-cycle entry and expansion in HIV-1 infection
    • Sereti I, Dunham RM, Spritzler J, IL-7 administration drives T cell-cycle entry and expansion in HIV-1 infection. Blood 2009; 113: 6304–6314
    • (2009) Blood , vol.113 , pp. 6304-6314
    • Sereti, I.1    Dunham, R.M.2    Spritzler, J.3
  • 125
    • 74549117108 scopus 로고    scopus 로고
    • Phase I study of recombinant human Interleukin-7 administration in subjects with refractory malignancy
    • Sportès C, Babb RR, Krumlauf MC, Phase I study of recombinant human Interleukin-7 administration in subjects with refractory malignancy. Clin Cancer Res 2010; 16: 727–735
    • (2010) Clin Cancer Res , vol.16 , pp. 727-735
    • Sportès, C.1    Babb, R.R.2    Krumlauf, M.C.3
  • 126
    • 33746046073 scopus 로고    scopus 로고
    • IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells
    • Rosenberg SA, Sportès C, Ahmadzadeh M, IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells. J Immunother 2006; 29: 313–319
    • (2006) J Immunother , vol.29 , pp. 313-319
    • Rosenberg, S.A.1    Sportès, C.2    Ahmadzadeh, M.3
  • 127
    • 79959551122 scopus 로고    scopus 로고
    • Thymosin-alpha1 promotes the apoptosis of regulatory T cells and survival rate in septic mice
    • Wan J, Shan Y, Shan H, Thymosin-alpha1 promotes the apoptosis of regulatory T cells and survival rate in septic mice. Front Biosci 2011; 16: 3004–3013
    • (2011) Front Biosci , vol.16 , pp. 3004-3013
    • Wan, J.1    Shan, Y.2    Shan, H.3
  • 128
    • 0022218775 scopus 로고
    • Modulation of natural killer activity by thymosin alpha 1 and interferon
    • Favalli C, Jezzi T, Mastino A, Modulation of natural killer activity by thymosin alpha 1 and interferon. Cancer Immunol Immunother 1985; 20: 189–192
    • (1985) Cancer Immunol Immunother , vol.20 , pp. 189-192
    • Favalli, C.1    Jezzi, T.2    Mastino, A.3
  • 129
    • 84957789576 scopus 로고    scopus 로고
    • Dual effect of Thymosin α 1 on human monocyte-derived dendritic cell in vitro stimulated with viral and bacterial toll-like receptor agonists
    • Giacomini E, Severa M, Cruciani M, Dual effect of Thymosin α 1 on human monocyte-derived dendritic cell in vitro stimulated with viral and bacterial toll-like receptor agonists. Expert Opin Biol Ther 2015; 15(Suppl 1): S59–S70
    • (2015) Expert Opin Biol Ther , vol.15 , pp. S59-S70
    • Giacomini, E.1    Severa, M.2    Cruciani, M.3
  • 130
    • 84893045720 scopus 로고    scopus 로고
    • Thymosin a1 activates complement receptor-mediated phagocytosis in human monocyte-derived macrophages
    • Serafino A, Pica F, Andreola F, Thymosin a1 activates complement receptor-mediated phagocytosis in human monocyte-derived macrophages. J Innate Immun 2014; 6: 72–88
    • (2014) J Innate Immun , vol.6 , pp. 72-88
    • Serafino, A.1    Pica, F.2    Andreola, F.3
  • 131
    • 84965087784 scopus 로고    scopus 로고
    • Thymosin alpha-1 treatment in chronic hepatitis B
    • Wu X, Jia J, You H. Thymosin alpha-1 treatment in chronic hepatitis B. Expert Opin Biol Ther 2015; 15(Suppl 1): S129–S132
    • (2015) Expert Opin Biol Ther , vol.15 , pp. S129-S132
    • Wu, X.1    Jia, J.2    You, H.3
  • 132
    • 17344368726 scopus 로고    scopus 로고
    • Combination therapy with thymosin alpha1 and interferon for the treatment of chronic hepatitis C infection: a randomized, placebo-controlled double-blind trial
    • Sherman KE, Sjogren M, Creager RL, Combination therapy with thymosin alpha1 and interferon for the treatment of chronic hepatitis C infection: a randomized, placebo-controlled double-blind trial. Hepatology 1998; 27: 1128–1135
    • (1998) Hepatology , vol.27 , pp. 1128-1135
    • Sherman, K.E.1    Sjogren, M.2    Creager, R.L.3
  • 133
    • 0035663014 scopus 로고    scopus 로고
    • The efficacy of thymosin in the treatment of chronic hepatitis B virus infection: a meta-analysis
    • Chan HLY, Tang JL, Tam W, The efficacy of thymosin in the treatment of chronic hepatitis B virus infection: a meta-analysis. Aliment Pharmol Ther 2001; 15: 1899–1905
    • (2001) Aliment Pharmol Ther , vol.15 , pp. 1899-1905
    • Chan, H.L.Y.1    Tang, J.L.2    Tam, W.3
  • 134
    • 84965089221 scopus 로고    scopus 로고
    • Historical review of thymosin α 1 in infectious diseases
    • Camerini R, Garaci E. Historical review of thymosin α 1 in infectious diseases. Expert Opin Biol Ther 2015; 15(Suppl 1): S117–S127
    • (2015) Expert Opin Biol Ther , vol.15 , pp. S117-S127
    • Camerini, R.1    Garaci, E.2
  • 135
    • 84987924231 scopus 로고    scopus 로고
    • The efficacy of thymosin α1 as immunomodulatory treatment for sepsis: a systematic review of randomized controlled trials
    • Liu F, Wang H-M, Wang T, The efficacy of thymosin α1 as immunomodulatory treatment for sepsis: a systematic review of randomized controlled trials. BMC Infect Dis 2016; 16: 488
    • (2016) BMC Infect Dis , vol.16 , pp. 488
    • Liu, F.1    Wang, H.-M.2    Wang, T.3
  • 136
    • 84872263573 scopus 로고    scopus 로고
    • The efficacy of thymosin alpha 1 for severe sepsis (ETASS): a multicenter, single-blind, randomized and controlled trial
    • Wu J, Zhou L, Liu J, The efficacy of thymosin alpha 1 for severe sepsis (ETASS): a multicenter, single-blind, randomized and controlled trial. Crit Care 2013; 17: R8
    • (2013) Crit Care , vol.17 , pp. R8
    • Wu, J.1    Zhou, L.2    Liu, J.3
  • 137
    • 0027443456 scopus 로고
    • The inhibitory actions of protease inhibitors on the production of polymorphonuclear leukocyte elastase and interleukin 8
    • Endo S, Inada K, Yamashita H, The inhibitory actions of protease inhibitors on the production of polymorphonuclear leukocyte elastase and interleukin 8. Res Commun Chem Pathol Pharmacol 1993; 82: 27–34
    • (1993) Res Commun Chem Pathol Pharmacol , vol.82 , pp. 27-34
    • Endo, S.1    Inada, K.2    Yamashita, H.3
  • 138
    • 77952492404 scopus 로고    scopus 로고
    • Urinary trypsin inhibitor as a therapeutic option for endotoxin-related inflammatory disorders
    • Inoue K-I, Takano H. Urinary trypsin inhibitor as a therapeutic option for endotoxin-related inflammatory disorders. Expert Opin Investig Drugs 2010; 19: 513–520
    • (2010) Expert Opin Investig Drugs , vol.19 , pp. 513-520
    • Inoue, K.-I.1    Takano, H.2
  • 139
    • 85008655979 scopus 로고    scopus 로고
    • Effect of ulinastatin combined with thymosin alpha1 on sepsis: A systematic review and meta-analysis of Chinese and Indian patients
    • Liu D, Yu Z, Yin J, Effect of ulinastatin combined with thymosin alpha1 on sepsis: A systematic review and meta-analysis of Chinese and Indian patients. J Crit Care 2017; 39: 259–266
    • (2017) J Crit Care , vol.39 , pp. 259-266
    • Liu, D.1    Yu, Z.2    Yin, J.3
  • 140
    • 84858394440 scopus 로고    scopus 로고
    • Bench-to-bedside review: Immunoglobulin therapy for sepsis – biological plausibility from a critical care perspective
    • Shankar-Hari M, Spencer J, Sewell WA, Bench-to-bedside review: Immunoglobulin therapy for sepsis – biological plausibility from a critical care perspective. Crit Care 2012; 16: 206
    • (2012) Crit Care , vol.16 , pp. 206
    • Shankar-Hari, M.1    Spencer, J.2    Sewell, W.A.3
  • 141
    • 85009740698 scopus 로고    scopus 로고
    • Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016
    • Rhodes A, Evans LE, Alhazzani W, Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Intensive Care Med 2017; 43: 304–377
    • (2017) Intensive Care Med , vol.43 , pp. 304-377
    • Rhodes, A.1    Evans, L.E.2    Alhazzani, W.3
  • 142
    • 84983196444 scopus 로고    scopus 로고
    • Intravenous immunoglobulin in septic shock: review of the mechanisms of action and meta-analysis of the clinical effectiveness
    • Busani S, Damiani E, Cavazzuti I, Intravenous immunoglobulin in septic shock: review of the mechanisms of action and meta-analysis of the clinical effectiveness. Minerva Anestesiol 2016; 82: 559–572
    • (2016) Minerva Anestesiol , vol.82 , pp. 559-572
    • Busani, S.1    Damiani, E.2    Cavazzuti, I.3
  • 143
    • 84918546294 scopus 로고    scopus 로고
    • Early therapy with IgM-enriched polyclonal immunoglobulin in patients with septic shock
    • Cavazzuti I, Serafini G, Busani S, Early therapy with IgM-enriched polyclonal immunoglobulin in patients with septic shock. Intensive Care Med 2014; 40: 1888–1896
    • (2014) Intensive Care Med , vol.40 , pp. 1888-1896
    • Cavazzuti, I.1    Serafini, G.2    Busani, S.3
  • 144
    • 84927768670 scopus 로고    scopus 로고
    • Intravenous immunoglobulin for severe sepsis and septic shock: clinical effectiveness, cost-effectiveness and value of a further randomised controlled trial
    • Soares MO, Welton NJ, Harrison DA, Intravenous immunoglobulin for severe sepsis and septic shock: clinical effectiveness, cost-effectiveness and value of a further randomised controlled trial. Crit Care 2014; 18: 649
    • (2014) Crit Care , vol.18 , pp. 649
    • Soares, M.O.1    Welton, N.J.2    Harrison, D.A.3
  • 145
    • 0025117997 scopus 로고
    • hemofiltration reverses left ventricular dysfunction during sepsis in dogs
    • Gomez A, Wang R, Unruh H, hemofiltration reverses left ventricular dysfunction during sepsis in dogs. Anesthesiology 1990; 73: 671–685
    • (1990) Anesthesiology , vol.73 , pp. 671-685
    • Gomez, A.1    Wang, R.2    Unruh, H.3
  • 146
    • 0034222937 scopus 로고    scopus 로고
    • Effects of different doses in continuous veno-venous haemofiltration on outcomes of acute renal failure: a prospective randomised trial
    • Ronco C, Bellomo R, Homel P, Effects of different doses in continuous veno-venous haemofiltration on outcomes of acute renal failure: a prospective randomised trial. Lancet 2000; 356: 26–30
    • (2000) Lancet , vol.356 , pp. 26-30
    • Ronco, C.1    Bellomo, R.2    Homel, P.3
  • 147
    • 33646416536 scopus 로고    scopus 로고
    • High-volume hemofiltration as salvage therapy in severe hyperdynamic septic shock
    • Cornejo R, Downey P, Castro R, High-volume hemofiltration as salvage therapy in severe hyperdynamic septic shock. Intensive Care Med 2006; 32: 713–722
    • (2006) Intensive Care Med , vol.32 , pp. 713-722
    • Cornejo, R.1    Downey, P.2    Castro, R.3
  • 148
    • 0033710084 scopus 로고    scopus 로고
    • Prospective evaluation of short-term, high-volume isovolemic hemofiltration on the hemodynamic course and outcome in patients with intractable circulatory failure resulting from septic shock
    • Honore PM, Jamez J, Wauthier M, Prospective evaluation of short-term, high-volume isovolemic hemofiltration on the hemodynamic course and outcome in patients with intractable circulatory failure resulting from septic shock. Crit Care Med 2000; 28: 3581
    • (2000) Crit Care Med , vol.28 , pp. 3581
    • Honore, P.M.1    Jamez, J.2    Wauthier, M.3
  • 149
    • 84941907184 scopus 로고    scopus 로고
    • Novel blood purification system for regulating excessive immune reactions in severe sepsis and septic shock: an ex vivo pilot study
    • Hara Y, Shimomura Y, Nakamura T, Novel blood purification system for regulating excessive immune reactions in severe sepsis and septic shock: an ex vivo pilot study. Ther Apher Dial 2015; 19: 308–315
    • (2015) Ther Apher Dial , vol.19 , pp. 308-315
    • Hara, Y.1    Shimomura, Y.2    Nakamura, T.3
  • 150
    • 1542719614 scopus 로고    scopus 로고
    • Hemoadsorption removes tumor necrosis factor, interleukin-6, and interleukin-10, reduces nuclear factor-kappaB DNA binding, and improves short-term survival in lethal endotoxemia
    • Kellum JA, Song M, Venkataraman R. Hemoadsorption removes tumor necrosis factor, interleukin-6, and interleukin-10, reduces nuclear factor-kappaB DNA binding, and improves short-term survival in lethal endotoxemia. Crit Care Med 2004; 32: 801–805
    • (2004) Crit Care Med , vol.32 , pp. 801-805
    • Kellum, J.A.1    Song, M.2    Venkataraman, R.3
  • 151
    • 42949146538 scopus 로고    scopus 로고
    • Effects of hemoadsorption on cytokine removal and short-term survival in septic rats
    • Peng Z-Y, Carter MJ, Kellum JA. Effects of hemoadsorption on cytokine removal and short-term survival in septic rats. Crit Care Med 2008; 36: 1573–1577
    • (2008) Crit Care Med , vol.36 , pp. 1573-1577
    • Peng, Z.-Y.1    Carter, M.J.2    Kellum, J.A.3
  • 152
    • 0027385965 scopus 로고
    • Experimental study of extracorporeal perfusion for septic shock
    • Sato T, Orlowski JP, Zborowski M. Experimental study of extracorporeal perfusion for septic shock. ASAIO J 1993; 39: M790–M793
    • (1993) ASAIO J , vol.39 , pp. M790-M793
    • Sato, T.1    Orlowski, J.P.2    Zborowski, M.3
  • 153
    • 67649219652 scopus 로고    scopus 로고
    • Early use of polymyxin B hemoperfusion in abdominal septic shock: the EUPHAS randomized controlled trial
    • Cruz DN, Antonelli M, Fumagalli R, Early use of polymyxin B hemoperfusion in abdominal septic shock: the EUPHAS randomized controlled trial. JAMA 2009; 301: 2445–2452
    • (2009) JAMA , vol.301 , pp. 2445-2452
    • Cruz, D.N.1    Antonelli, M.2    Fumagalli, R.3
  • 154
    • 84931957864 scopus 로고    scopus 로고
    • Early use of polymyxin B hemoperfusion in patients with septic shock due to peritonitis: a multicenter randomized control trial
    • Payen DM, Guilhot J, Launey Y, Early use of polymyxin B hemoperfusion in patients with septic shock due to peritonitis: a multicenter randomized control trial. Intensive Care Med 2015; 41: 975–984
    • (2015) Intensive Care Med , vol.41 , pp. 975-984
    • Payen, D.M.1    Guilhot, J.2    Launey, Y.3
  • 155
    • 84981719229 scopus 로고    scopus 로고
    • Polymyxin-B hemoperfusion in septic patients: analysis of a multicenter registry
    • Cutuli SL, Artigas A, Fumagalli R, Polymyxin-B hemoperfusion in septic patients: analysis of a multicenter registry. Ann Intensive Care 2016; 6: 77
    • (2016) Ann Intensive Care , vol.6 , pp. 77
    • Cutuli, S.L.1    Artigas, A.2    Fumagalli, R.3
  • 156
    • 84903130991 scopus 로고    scopus 로고
    • The EUPHRATES trial (Evaluating the Use of Polymyxin B Hemoperfusion in a Randomized controlled trial of Adults Treated for Endotoxemia and Septic shock): study protocol for a randomized controlled trial
    • Klein DJ, Foster D, Schorr CA, The EUPHRATES trial (Evaluating the Use of Polymyxin B Hemoperfusion in a Randomized controlled trial of Adults Treated for Endotoxemia and Septic shock): study protocol for a randomized controlled trial. Trials 2014; 15: 218
    • (2014) Trials , vol.15 , pp. 218
    • Klein, D.J.1    Foster, D.2    Schorr, C.A.3
  • 157
    • 85021669734 scopus 로고    scopus 로고
    • Is polymyxin B-immobilized fiber column ineffective for septic shock? A discussion on the press release for EUPHRATES trial
    • Iba T, Fowler L. Is polymyxin B-immobilized fiber column ineffective for septic shock? A discussion on the press release for EUPHRATES trial. J Intensive Care 2017; 5: 40
    • (2017) J Intensive Care , vol.5 , pp. 40
    • Iba, T.1    Fowler, L.2
  • 158
    • 85016153039 scopus 로고    scopus 로고
    • Hemoadsorption by CytoSorb in septic patients: a case series
    • Kogelmann K, Jarczak D, Scheller M, Hemoadsorption by CytoSorb in septic patients: a case series. Crit Care 2017; 21: 74
    • (2017) Crit Care , vol.21 , pp. 74
    • Kogelmann, K.1    Jarczak, D.2    Scheller, M.3
  • 159
    • 84911445227 scopus 로고    scopus 로고
    • An extracorporeal blood-cleansing device for sepsis therapy
    • Kang JH, Super M, Yung CW, An extracorporeal blood-cleansing device for sepsis therapy. Nat Med 2014; 20: 1211–1216
    • (2014) Nat Med , vol.20 , pp. 1211-1216
    • Kang, J.H.1    Super, M.2    Yung, C.W.3
  • 160
    • 84939621978 scopus 로고    scopus 로고
    • Improved treatment of systemic blood infections using antibiotics with extracorporeal opsonin hemoadsorption
    • Didar TF, Cartwright MJ, Rottman M, Improved treatment of systemic blood infections using antibiotics with extracorporeal opsonin hemoadsorption. Biomaterials 2015; 67: 382–392
    • (2015) Biomaterials , vol.67 , pp. 382-392
    • Didar, T.F.1    Cartwright, M.J.2    Rottman, M.3
  • 161
    • 0028354887 scopus 로고
    • Noradrenaline inhibits lipopolysaccharide-induced tumor necrosis factor and interleukin 6 production in human whole blood
    • van der Poll T, Jansen J, Endert E, Noradrenaline inhibits lipopolysaccharide-induced tumor necrosis factor and interleukin 6 production in human whole blood. Infect Immun 1994; 62: 2046–2050
    • (1994) Infect Immun , vol.62 , pp. 2046-2050
    • van der Poll, T.1    Jansen, J.2    Endert, E.3
  • 162
    • 77953285381 scopus 로고    scopus 로고
    • Noradrenaline acting at beta-adrenoceptors induces expression of IL-1beta and its negative regulators IL-1ra and IL-1RII, and drives an overall anti-inflammatory phenotype in rat cortex
    • McNamee EN, Griffin EW, Ryan KM, Noradrenaline acting at beta-adrenoceptors induces expression of IL-1beta and its negative regulators IL-1ra and IL-1RII, and drives an overall anti-inflammatory phenotype in rat cortex. Neuropharmacology 2010; 59: 37–48
    • (2010) Neuropharmacology , vol.59 , pp. 37-48
    • McNamee, E.N.1    Griffin, E.W.2    Ryan, K.M.3
  • 163
    • 23044490894 scopus 로고    scopus 로고
    • Norepinephrine, dopamine and dexamethasone modulate discrete leukocyte subpopulations and cytokine profiles from human PBMC
    • Torres KCL, Antonelli LRV, Souza ALS, Norepinephrine, dopamine and dexamethasone modulate discrete leukocyte subpopulations and cytokine profiles from human PBMC. J Neuroimmunol 2005; 166: 144–157
    • (2005) J Neuroimmunol , vol.166 , pp. 144-157
    • Torres, K.C.L.1    Antonelli, L.R.V.2    Souza, A.L.S.3
  • 164
    • 84879755954 scopus 로고    scopus 로고
    • Norepinephrine inhibits macrophage migration by decreasing CCR2 expression
    • Xiu F, Stanojcic M, Jeschke MG. Norepinephrine inhibits macrophage migration by decreasing CCR2 expression. PLoS One 2013; 8: e69167
    • (2013) PLoS One , vol.8 , pp. e69167
    • Xiu, F.1    Stanojcic, M.2    Jeschke, M.G.3
  • 165
    • 70449083312 scopus 로고    scopus 로고
    • Vasopressin attenuates TNF-mediated inflammation in the rat cremaster microcirculation
    • McMahon PJ, Proctor KG. Vasopressin attenuates TNF-mediated inflammation in the rat cremaster microcirculation. J Trauma 2009; 67: 461–473
    • (2009) J Trauma , vol.67 , pp. 461-473
    • McMahon, P.J.1    Proctor, K.G.2
  • 166
    • 54249165079 scopus 로고    scopus 로고
    • Vasopressin decreases sepsis-induced pulmonary inflammation through the V2R
    • Boyd JH, Holmes CL, Wang Y, Vasopressin decreases sepsis-induced pulmonary inflammation through the V2R. Resuscitation 2008; 79: 325–331
    • (2008) Resuscitation , vol.79 , pp. 325-331
    • Boyd, J.H.1    Holmes, C.L.2    Wang, Y.3
  • 167
    • 84883790310 scopus 로고    scopus 로고
    • Vasopressin inhibits endotoxin-induced upregulation of inflammatory mediators in activated macrophages
    • Peng T-C, Huang C-J. Vasopressin inhibits endotoxin-induced upregulation of inflammatory mediators in activated macrophages. Tzu Chi Med J 2013; 25: 150–154
    • (2013) Tzu Chi Med J , vol.25 , pp. 150-154
    • Peng, T.-C.1    Huang, C.-J.2
  • 168
    • 85058429039 scopus 로고    scopus 로고
    • Vasopressin alone and with noradrenaline attenuates TNF-α production in an in-vitro model of monocyte priming and deactivation
    • Davies R, O' Dea KP, Soni S, Vasopressin alone and with noradrenaline attenuates TNF-α production in an in-vitro model of monocyte priming and deactivation. Crit Care 2017; 21: P362
    • (2017) Crit Care , vol.21 , pp. P362
    • Davies, R.1    O' Dea, K.P.2    Soni, S.3
  • 169
    • 84881173520 scopus 로고    scopus 로고
    • Vasopressin compared with norepinephrine augments the decline of plasma cytokine levels in septic shock
    • Russell JA, Fjell C, Hsu JL, Vasopressin compared with norepinephrine augments the decline of plasma cytokine levels in septic shock. Am J Respir Crit Care Med 2013; 188: 356–364
    • (2013) Am J Respir Crit Care Med , vol.188 , pp. 356-364
    • Russell, J.A.1    Fjell, C.2    Hsu, J.L.3
  • 170
    • 84982132621 scopus 로고    scopus 로고
    • Effect of early vasopressin vs norepinephrine on kidney failure in patients with septic shock: The VANISH Randomized Clinical Trial
    • Gordon AC, Mason AJ, Thirunavukkarasu N, Effect of early vasopressin vs norepinephrine on kidney failure in patients with septic shock: The VANISH Randomized Clinical Trial. JAMA 2016; 316: 509–518
    • (2016) JAMA , vol.316 , pp. 509-518
    • Gordon, A.C.1    Mason, A.J.2    Thirunavukkarasu, N.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.