Anemia of inflammation: features, the necessity for and possibility of correction (literature review)
#2019-1
PDF_2019-01_20-35 (Русский)

Keywords

anemia of inflammation
iron
iron metabolism

How to Cite

Orlov Y.P., Govorova N.V., Nochnaja Y.A., Rudnov V.A. Anemia of inflammation: features, the necessity for and possibility of correction (literature review). Annals of Critical Care. 2019;(1):20–35. doi:10.21320/1818-474X-2019-1-20-35.

Statistic

Abstract Views: 1584
PDF_2019-01_20-35 (Русский) Downloads: 184
Statistic from 21.01.2023

Abstract

The purpose for writing this review. Analysis of publications in search engines about physiological and pathological iron metabolism, pathogenesis of anemia of inflammation that develops in patients in the ICU, to determine possible indications and contraindications for therapeutic intervention.

Methods. Analyzed articles in the medical literature databases, Pubmed, Medline, and EMBASE. The search strategy used for keyword: “anemia of inflammation”, “iron and infection, anaemia and sepsis”, “free hemoglobin, iron Exchange” in the period from 1990 up to 2018, inclusive and affordable domestic work (e-library) literature. The materials used by the worldʼs leading organizations World Health Organization, the Cochrane Reviews, WSACS, ARDS ARTS CENTRE Clinical Trials Network, European Society of Intensive Care Medicine, European Society of Anesthesiologists, Society of Critical Care Medicine.

Conclusion. Anemia as a symptom, in critical condition in the first place, requires a definition of its role in the genesis of hypoxia. This should be supported by not only hemoglobin levels, as a carrier of oxygen and hypoxia specific criteria: analysis of blood gases, assess the height of tooth ST according to ECG and, of course, blood lactate level (more than 2 mmol/l). Anemia when sepsis is caused by intravascular hemolysis and gipoferremija is a consequence of natural compensatory protection from possible manifestations of infection. The introduction of such patients with iron or blood donations entail access to iron bacteria. In the context of sepsis and continuing to use the hemolysis chelators, if revealed high levels of ferritin, low concentration of gaptoglobina and transferrin. Decision on blood transfusion or iron supplementation should be individualized, taking into account the specific factors of the patient, and any potential benefits of therapy drugs iron should.

https://doi.org/10.21320/1818-474X-2019-1-20-35
PDF_2019-01_20-35 (Русский)

Full-text of the article is available for this locale: Русский.

References

  1. Poggiali E., Migone De Amicis M., Motta I. Anemia of chronic disease: a unique defect of iron recycling for many different chronic diseases. Eur. J. Intern. Med. 2014; 25(1): 12–7. DOI: 10.1111/ggi.12371:10.1016/j.ejim
  2. Corwin H.L., Gettinger A., Pearl R.G., et al. The CRIT Study: Anemia and blood transfusion in the critically ill-current clinical practice in the United States. Crit. Care Med. 2004;32(1): 39–52. DOI: 10.1097/01.CCM.0000104112.34142.79
  3. Joosten E., Lioen P. Iron deficiency anemia and anemia of chronic disease in geriatric hospitalized patients: How frequent are comorbidities as an additional explanation for the anemia? Geriatr Gerontol Int. 2015; 15(8): 931–935. DOI: 10.1111/ggi.12371
  4. Cartwright G.E. The anemia of chronic disorders. Semin Hematol. 1966; 3: 351–375.
  5. Zhang D.L., Ghosh M.C., Rouault T.A. The physiological functions of iron regulatory proteins in iron homeostasis — an update. Front. Pharmacol. 2014; 5: 124. DOI: 10.3389/fphar.2014.00124
  6. Andrews N.C., Schmidt P.J. Iron homeostasis. Annu Rev. Physiol. 2007; 69: 69–85. DOI: 10.1146/annurev.physiol.69.031905.164337
  7. Pantopoulos K., Porwal S.K., Tartakoff A., Devireddy L. Mechanisms of mammalian iron homeostasis. Send to Biochemistry. 2012; 51(29): 5705–5724. DOI: 10.1021/bi300752r
  8. Schaer D.J, Buehler P.W., Alayash A.I, et al. Positive Iron Balance in Chronic Kidney Disease: How Much is Too Much and How to Tell? Send to Am. J. Nephrol. 2018; 47(2): 72–83. DOI: 10.1159/000486968
  9. Philpott C.C., Jadhav S. The ins and outs of iron: Escorting iron through the mammalian cytosol. Free Radic Biol. Med. 2018; S0891–5849(18)32167–1. DOI: 10.1016/j.freeradbiomed.2018.10.411
  10. Gozzelino R., Arosio P. Iron Homeostasis in Health and Disease. Int. J. Mol. Sci. 2016; 17(1): 130. DOI: 10.3390/ijms17010130
  11. Bullen J.J. The signifi cance of iron in infection. Rev. Infect. Dis. 1981; 3: 1127–1138.
  12. Зайчик А.Ш., Чурилов Л.П. Основы патохимии. Учебник для студентов медицинских вузов. СПб.: ЭЛБИ-СПб. 2001: 688.
  13. [Zajchik A.Sh., CHurilov L.P. Osnovy patohimii. Uchebnik dlya studentov medicinskih vuzov). SPb.: EHLBI-SPb. 2001: 688. (In Russ)]
  14. Abbaspour N., Hurrell R., Kelishadi R. Review on iron and its importance for human health. J. Res. Med. Sci. 2014; 19(2): 164–174. PMCID PMC3999603
  15. Belcher J.D., Beckman J.D., Balla G., Balla J., Vercellotti G. Heme degradation and vascular injury. DOI: 10.1089/ars.2009.2822
  16. Park C.H., Valore E.V., Waring A.J., Ganz T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J. Biol. Chem. 2001; 276: 7806–7810. DOI: 10.1074/jbc.M008922200
  17. Krause A., Neitz S., Mägert H.J., et al. LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity. FEBS Lett. 2000; 480: 147–150. DOI.org/10.1016/S0014–5793(00)01920–7
  18. Finberg K.E. Unraveling mechanisms regulating systemic iron homeostasis. Hematology Am. Soc. Hematol. Educ. Program. 2011; 2011:532–537. DOI: 10.1182/asheducation-2011.1.532
  19. Martins A.C., Almeida J.I., Lima I.S., et al. Iron Metabolism and the Inflammatory Response. DOI: 10.1002/iub.1635
  20. Madua Anazoeze J., Ughasoro Maduka D. Anaemia of Chronic Disease: An In-Depth Review. DOI: 10.1159/000452104
  21. Kell D.B. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med. Genomics. 2009; 2: 2. DOI: 10.1186/1755-8794-2-2
  22. Wardman P., Candeias L.P. Fenton chemistry: An introduction. Rad. Res. 1996; 145: 523–531.
  23. Kehrer J.P. The Haber-Weiss reaction and mechanisms of toxicity. Toxicology. 2000; 149: 43–50. PMID 10963860
  24. Nadadur S.S., Srirama K., Mudipalli A. Iron transport & homeostasis mechanisms: their role in health & disease. Indian J. Med. Res. 2008;128(4): 533–544. PMID 19 106445
  25. Kruzel M.L., Zimecki M., Actor J.K. Lactoferrin in a Context of Inflammation-Induced Pathology. Front Immunol. 2017; 8: 1438. DOI: 10.3389/fimmu.
  26. Dutra F.F., Bozza M.T. Heme on innate immunity and inflammation. Front Pharmacol. 2014; 5: 115. DOI: 10.3389/fphar.
  27. Belcher J.D., Chen C., Nguyen J., et al. Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease. Blood. 2014: 123377–390. DOI: 10.1182/blood-2013-04-495887
  28. Gozzelino R., Jeney V., Soares M.P. Mechanisms of cell protection by heme oxygenase-1. Annu Rev. Pharmacol Toxicol. 2010; 50: 323–354. DOI: 10.1146/annurev.pharmtox.010909.105600
  29. Larsen R., Gozzelino R., Jeney V., et al. A central role for free heme in the pathogenesis of severe sepsis. Sci Transl. Med. 2010; 2(51): 51ra71. DOI: 10.1126/scitranslmed.3001118
  30. Иванов А.В., Долгих В.Т., Лукач В.Н., Орлов Ю.П. Критические состояния как логическая и закономерная цепь событий в нарушении метаболизма железа (обобщение экспериментальных исследований). Биомедицинская химия. 2013; 59(6): 700–709.
  31. [Ivanov A.V., Dolgih V.T., Lukach V.N., Orlov Yu.P. Kriticheskie sostoyaniya kak logicheskaya i zakonomernaya cepʼ sobytij v narushenii metabolizma zheleza (obobshchenie ehksperimentalʼnyh issledovanij). Biomedicinskaya himiya. 2013; 59(6): 700–709. (In Russ)]
  32. Vinchi F., Tolosano E. Therapeutic approaches to limit hemolysis-driven endothelial dysfunction: scavenging free heme to preserve vasculature homeostasis. Oxid Med. Cell. Longev. 2013; 2013: 396527. DOI: 10.1155/2013/396527
  33. Li S., Fujino M., Takahara T., Li X.K. Protective role of heme oxygenase-1 in fatty liver ischemia-reperfusion injury. Med. Mol. Morphol. 2018; Aug 31. DOI: 10.1007/s00795-018-0204-0
  34. Zhang F.H., Sun Y.H., Fan K.L., et al. Protective effects of heme oxygenase-1 against severe acute pancreatitis via inhibition of tumor necrosis factor-α and augmentation of interleukin-10. BMC Gastroenterol. 2017; 17(1): 100. DOI: 10.1186/s12876-017-0651-4
  35. Орлов Ю.П., Лукач В.Н., Долгих В.Т., Соболева Е.Л., Иванова А.М. Роль ионов железа в нарушении микроциркуляции и реологических свойств крови при ишемии/реперфузии в эксперименте. Вестник анестезиологии и реаниматологии. 2012; 9(3): 51–54.
  36. [Orlov Yu.P., Lukach V.N., Dolgih V.T., Soboleva E.L., Ivanova A.M. Rolʼ ionov zheleza v narushenii mikrocirkulyacii i reologicheskih svojstv krovi pri ishemii/reperfuzii v ehksperimente. Vestnik anesteziologii i reanimatologii. 2012; 9(3): 51–54. (In Russ)]
  37. Мчедлишвили Г.И. Гемореология в системе микроциркуляции: ее специфика и практическое значение. Тромбоз, гемостаз и реология. 2002; 4(12): 18–24.
  38. [Mchedlishvili G.I. Gemoreologiya v sisteme mikrocirkulyacii: ee specifika i prakticheskoe znachenie. Tromboz, gemostaz i reologiya. 2002; 4(12): 18–24. (In Russ)]
  39. Шидловский А.С., Салтанов А.И. Варианты механизмов изменения активности трансаминаз: клиническая интерпретация. Вестник интенсивной терапии, 2015, 1: 22–32.
  40. [Shidlovskij A.S., Saltanov A.I. Varianty mekhanizmov izmeneniya aktivnosti transaminaz: klinicheskaya interpretaciya. Vestnik intensivnoj terapii, 2015, 1: 22–32. (In Russ)]
  41. Effenberger-Neidnicht K., Hartmann M. Mechanisms of Hemolysis During Sepsis. Send to Inflammation. 2018; 41(5): 1569–1581. DOI: 10.1007/s10753-018-0810-y
  42. Brauckmann S., Effenberger-Neidnicht K., de Groot H., et al. Lipopolysaccharide-induced hemolysis: Evidence for direct membrane interactions. Sci Rep. 2016; 6: 35508. DOI: 10.1038/srep35508
  43. Gomes A.C., Moreira A.C., Mesquita G., Gomes M.S. Modulation of Iron Metabolism in Response to Infection: Twists for All Tastes. Send to Pharmaceuticals (Basel). 2018; 11(3): E84. DOI: 10.3390/ph11030084
  44. Butt A.T., Thomas M.S. Iron Acquisition Mechanisms and Their Role in the Virulence of Burkholderia Species. Front. Cell. Infect. Microbiol. 2017; 7: 460. DOI: 10.3389/fcimb.2017.00460
  45. Ganz T. Iron and infection. Int. J. Hematol. 2018; 107(1): 7–15. DOI: 10.1007/s12185-017-2366-2. Epub 2017 Nov 16
  46. Barber M.F., Elde N.C. Buried treasure: evolutionary perspectives on microbial iron piracy. Trends Genet. 2015; 31: 627–36. DOI: 10.1016/j.tig.2015.09.001
  47. Бухарин О.В., Усвяцов Б.Я., Щуплова Е.А. Антигемоглобиновая активность бактерий при взаимодействии с эритроцитами и ее роль в патогенезе анемии. Журнал микробиологии, эпидемиологии и иммунобиологии. 2011; 4: 25–29.
  48. [Buharin O.V., Usvyacov B.Ya., Shchuplova E.A. Antigemoglobinovaya aktivnostʼ bakterij pri vzaimodejstvii s ehritrocitami i ee rolʼ v patogeneze anemii. Zhurnal mikrobiologii, ehpidemiologii i immunobiologii. 2011; 4: 25–29. (In Russ)]
  49. Yamaguchi M., Terao Y., Mori-Yamaguchi Y., et al. Streptococcus pneumoniae invades erythrocytes and utilizes them to evade human innate immunity. PLoS One. 2013; 8(10): e77282. DOI: 10.1371/journal.pone.0077282
  50. Weiskopf R.B., Viele M.K., Feiner J., et al. Human cardiovascular and metabolic response to acute, severe isovolemic anemia. JAMA. 1998; 279(3): 217–221.
  51. Semenza G.L. Regulation of oxygen homeostasis by hypoxia-inducible factor 1. Physiology (Bethesda). 2009;24: 97–106. DOI: 10.1152/physiol.00045.2008
  52. Schönhofer B., Wenzel M., Geibel M., Köhler D. Blood transfusion and lung function in chronically anemic patients with severe chronic obstructive pulmonary disease. Crit. Care Med. 1998; 26: 1824–1828.
  53. Winslow R.M., Monge C.C., Brown E.G., et al. Effects of hemodilution on O2 transport in high-altitude polycythemia. J. Appl. Physiol. 1985; 59: 1495–1502.
  54. Cartwright G.E., Lauritsen M.A., Jones P.J., et al. The anemia of infection. I. Hypoferremia, hepercupremia, and alteration in porphyrin metabolism in patient. J. Clin Invest. 1946; 25: 65–80.
  55. Cartwright G.E., Lauritsen M.A., Humphreys S., et al. The anemia of infection. II. The experimental production of Hypoferremia and anemia in dogs. J. Clin. Invest. 1946; 25: 81–86.
  56. Besarab A., Frinak S., Yee J. An indistinct balance: The safety and efficacy of parenteral iron therapy. J. Am. Soc. Nephrol. 1999; 10: 2029–2043.
  57. Cieri E. Does iron cause bacterial infections in patients with end stage renal disease? ANNA J. 1999; 26: 591–596.
  58. Fishbane S. Review of issues relating to iron and infection. Am. J. Kidney Dis. 1999; 34(Suppl. 2): S47–S52.
  59. Hoen B. Iron and infection: Clinical experience. Am. J. Kidney Dis. 1999; 34(Suppl. 2): S30–S34.
  60. Patruta S.I., Hörl W.H. Iron and infection. Kidney Int. Suppl. 1999; 69: S125–S130.
  61. Holbein B.E. Iron-controlled infection with Neisseria meningitidis in mice. Infect Immun. 1980; 29: 886–891.
  62. Beaumier D.L., Caldwell M.A., Holbein B.E. Inflammation triggers hypoferremia and de novo synthesis of serum transferrin and ceruloplasmin in mice. Infect Immun. 1984; 46: 489–494.
  63. Bertini R., Bianchi M., Erroi A., et al. Dexamethasone modulation of in vivo effects of endotoxin, tumor necrosis factor, and interleukin-1 on liver cytochrome P-450, plasma fibrinogen, and serum iron. J. Leukoc. Biol. 1989; 46: 254–262.
  64. Schaible U.E., Collins H.L., Priem F., Kaufmann S.H. Correction of the iron overload defect in beta-2-microglobulin knockout mice by lactoferrin abolishes their increased susceptibility to tuberculosis. J. Exp. Med. 2002; 196: 1507–1513. PMCID PMC2194267
  65. Ganz T., Nemeth E. Iron sequestration and anemia of inflammation. Semin. Hematol. 2009; 46:387–393. DOI: 10.1053/j.seminhematol.2009.06.001
  66. Holbein B.E. Enhancement of Neisseria meningitidis infection in mice by addition of iron bound to transferrin. Infect Immun. 1981; 34: 120–125.
  67. Kemna E., Pickkers P., Nemeth E., van der Hoeven H., Swinkels D. Time-course analysis of hepcidin, serum iron, and plasma cytokine levels in humans injected with LPS. Blood. 2005; 106: 1864–1866. DOI: 10.1182/blood-2005-03-1159
  68. Fillet G., Cook J.D., Finch C.A. Storage iron kinetics. VII. A biologic model for reticuloendothelial iron transport. J. Clin. Invest. 1974; 53: 1527–1533.
  69. Noyes W.D., Bothwell T.H., Finch C.A. The role of the reticulo-endothelial cell in iron metabolism. Br. J. Haematol. 1960; 6: 43–55.
  70. Spitalnik S.L. Stored RBC Transfusions: Iron, Inflammation, Immunity, Infection 2013 Emily Cooley Lecture. Transfusion. 2014; 54(10): 2365–2371. DOI: 10.1111/trf.12848
  71. Freidank H.M., Billing H., Wiedmann-Al-Ahmad M. Influence of iron restriction on Chlamydia pneumoniae and C. trachomatis. Journal of medical microbiology. 2001; 50: 223–227. DOI: 10.1099/0022-1317-50-3-223
  72. Nairz M, et al. Genetic and Dietary Iron Overload Differentially Affect the Course of Salmonella Typhimurium Infection. Front. Cell. Infect. Microbiol. 2017; 7: 110. DOI: 10.3389/fcimb.2017.00110
  73. Prakash D. Anemia in the ICU: anemia of chronic disease versus anemia of acute illness. Crit. Care Clin. 2012; 28: 333–343. DOI: 10.1016/j.ccc.2012.04.012
  74. Pieracci F.M., Barie P.S. Diagnosis and management of iron-related anemias in critical illness. Crit. Care Med. 2006; 34: 1898–1905. DOI: 10.1097/01.CCM.0000220495.10510.C1
  75. Sihler K.C., Napolitano L.M. Anemia of inflammation in critically ill patients. J. Intensive Care Med. 2008; 23: 295–302. DOI: 10.1177/0885066608320836
  76. Piagnerelli M., Cotton F., Herpain A., et al. Time course of iron metabolism in critically ill patients. Acta Clin. Belg. 2013; 68(1): 22–27. DOI: 10.2143/ACB.68.1.2062715
  77. Nemeth E., Rivera S., Gabayan V., et al. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J. Clin. Invest. 2004; 113: 1271–1276. DOI: 10.1172/JCI20945
  78. Weiss G., Goodnough L.T. Anemia of chronic disease. N. Engl. J. Med. 2005; 352(10): 1011–1023. DOI: 10.1056/NEJMra041809
  79. Dinkla S., van Eijk L.T., Fuchs B., et al. Inflammation-associated changes in lipid composition and the organization of the erythrocyte membrane. BBA Clin. 2016; 5: 186–192. DOI: 10.1016/j.bbacli.2016.03.007
  80. Georgatzakou H.T., Antonelou M.H., Papassideri I.S., Kriebardis A.G. Red blood cell abnormalities and the pathogenesis of anemia in end-stage renal disease. Proteomics Clin. Appl. 2016; 10(8): 778–790. DOI: 10.1002/prca.201500127
  81. Brookhart M.A., Freburger J.K., Ellis A.R., et al. Infection Risk with Bolus versus Maintenance Iron Supplementation in Hemodialysis Patients. J. Am. Soc. Nephrol. 2013; 24(7): 1151–1158. DOI: 10.1681/ASN.2012121164
  82. Collins A.J., Ebben J., Ma J.Z., Xia H. Iron dosing patterns and mortality [Abstract] J. Am.Soc. Nephrol. 1998; 9: 250A.
  83. Кремлинг Х., Лутцайер В., Хайнтц Р. Гинекологическая урология и нефрология: пер. с нем. М.: Медицина, 1985.
  84. [Kremling H., Lutcajer V., Hajntc R. Ginekologicheskaya urologiya i nefrologiya: per. s nem. M.: Medicina, 1985. (In Russ)]
  85. Лыкова О.Ф., Захарова Е.В., Конышева Т.В., Хохлова З.А. Содержание лактоферрина в сыворотке крови и ликворе больных менингитом. Журнал микробиологии эпидемиологии и иммунобиологии. 2007; 2: 80–84.
  86. [Lykova O.F., Zaharova E.V., Konysheva T.V., Hohlova Z.A. Soderzhanie laktoferrina v syvorotke krovi i likvore bolʼnyh meningitom. Zhurnal mikrobiologii ehpidemiologii i immunobiologii. 2007; 2: 80–84. (In Russ)]
  87. Tacke F., Nuraldeen R., Koch A., et al. Iron Parameters Determine the Prognosis of Critically Ill Patients. Crit. Care Med. 2016; 44(6): 1049–1058. DOI: 10.1097/CCM.0000000000001607
  88. Lan P., Pan K.H., Wang S.J., et al. High Serum Iron level is Associated with Increased Mortality in Patients with Sepsis. Sci Rep. 2018; 8(1): 11072. DOI: 10.1038/s41598-018-29353-2
  89. Giraud B., Frasca D., Debaene B., et al. Comparison of haemoglobin measurement methods in the operating theatre. Br. J. Anaesth. 2013; 111: 946–54. DOI: 10.1093/bja/aet252
  90. Lelubre C., Vincent J.L. Red blood cell transfusion in the critically ill patient. Ann. Intensive Care. 2011; 1: 43. DOI: 10.1186/2110-5820-1-43
  91. Youssef L.A., Spitalnik S.L. Iron: a double-edged sword. Transfusion. 2017; 57(10): 2293–2297. DOI: 10.1111/trf.14296
  92. Weiskopf R.B., Kramer J.H., Viele M., et al. Acute severe isovolemic anemia impairs cognitive function and memory in humans. Anesthesiology. 2000; 92: 1646–1652.
  93. Vallet B., Robin E., Lebuffe G. Venous oxygen saturation as a physiologic transfusion trigger. Crit. Care. 2010; 14: 213. DOI: 10.1186/cc8854
  94. Yalavatti G.S., DeBacker D., Vincent J.L. Assessment of cardiac index in anemic patients. Chest. 2000; 118: 782–787.
  95. Hod E.A., Zhang N., Sokol S.A., et al. Transfusion of red blood cells after prolonged storage produces harmful effects that are mediated by iron and inflammation. Blood. 2010; 115(21): 4284–42892. DOI: 10.1182/blood-2009-10-245001
  96. Wang L., Johnson E.E., Shi H.N., et al. Attenuated inflammatory responses in hemochromatosis reveal a role for iron in the regulation of macrophage cytokine translation. J. Immunol. 2008; 181(4): 2723–2731. PMC 2561261
  97. Nixon A.M., Neely E., Simpson I.A., Connor J.R. The role of HFE genotype in macrophage phenotype. J. Neuroinflammation. 2018; 15(1): 30. DOI: 10.1186/s12974-018-1057-0.
  98. Gordeuk V.R., Ballou S., Lozanski G., Brittenham G.M. Decreased concentrations of tumor necrosis factor-alpha in supernatants of monocytes from homozygotes for hereditary hemochromatosis. Blood. 1992; 79(7): 1855–1860.
  99. von Bonsdorff L., Sahlstedt L., Ebeling F., et al. Apotransferrin administration prevents growth of Staphylococcus epidermidis in serum of stem cell transplant patients by binding of free iron. FEMS Immunol. Med. Microbiol. 2003; 37(1): 45–51. DOI: 10.1016/S0928–8244(03)00109–3
  100. Hebert P.C., Wells G., Blajchman M.A., et al. A multicenter, randomized, controlled clinical trial of transfusion requirements in critical care. N. Engl. J. Med. 1999; 340: 409–417.
  101. García-Roa M., Del Carmen Vicente-Ayuso M., Bobes A.M., et al. Review Red blood cell storage time and transfusion: current practice, concerns and future perspectives. Blood Transfus. 2017; 15(3): 222–231. DOI: 10.2450/2017.0345–16
  102. Litton E., Baker S., Erber W.N., et al. Intravenous iron or placebo for anaemia in intensive care: the IRONMAN multicentre randomized blinded trial: A randomized trial of IV iron in critical illness. Intensive Care Med. 2016; 42(11): 1715–1722. DOI: 10.1007/s00134-016-4465-6
  103. Garrido-Martín P., Nassar-Mansur M.I., de la Llana-Ducrós R., et al. The effect of intravenous and oral iron administration on perioperative anaemia and transfusion requirements in patients undergoing elective cardiac surgery: a randomized clinical trial. Interact Cardiovasc Thorac Surg. 2012; 15(6): 1013–1038; DOI: 10.1093/icvts/ivs344
  104. Pieracci F.M., Henderson P., Rodney J.R., et al. Randomized, double-blind, placebo-controlled trial of effects of enteral iron supplementation on anemia and risk of infection during surgical critical illness. Surg. Infect. (Larchmt). 2009; 10(1): 9–19. DOI: 10.1089/sur.2008.043
  105. Shah A., Roy N.B., McKechnie S., et al. Iron supplementation to treat anaemia in adult critical care patients: a systematic review and meta-analysis. Crit Care. 2016; 20: 306. DOI: 10.1186/s13054-016-1486-z
  106. Litton E., Xiao J., Ho K.M. Safety and efficacy on intravenous iron therapy in reducing requirement for allogeneic blood transfusion: a systematic review and meta-analysis of randomised clinical trials. BMJ. 2013; 347: f4822. DOI: 10.1136/bmj.f4822
  107. Pasricha S.R., Atkinson S.A., Armitage A.E., et al. Expression of the Iron Hormone Hepcidin Distinguishes Different Types of Anemia in African Children. Sci Transl. Med. 2014; 6: 235re3. DOI: 10.1126/scitranslmed.3008249
  108. Bregman D.B., Morris D., Koch T.A., et al. Hepcidin levels predict nonresponsiveness to oral iron therapy in patients with iron deficiency anaemia. Am. J. Hematol. 2013; 88: 97–101. DOI: 10.1002/ajh.23354
  109. Jelkmanna I., Jelkmannb W. Impact of Erythropoietin on Intensive Care Unit Patients. Transfus Med Hemother. 2013; 40(5): 310–318. DOI: 10.1159/000354128
  110. van Iperen C.E., Gaillard C.A., Kraaijenhagen R.J., et al. Response of erythropoiesis and iron metabolism to recombinant human erythropoietin in intensive care unit patients. Crit. Care Med. 2000; 28(8): 2773–2778.
  111. Vincent J.L., Spapen H.D., Creteur J., et al. Pharmacokinetics and pharmacodynamics of once-weekly subcutaneous epoetin alfa in critically ill patients: results of a randomized, double-blind, placebo-controlled trial. Crit. Care Med. 2006;34(6): 1661–1667. DOI: 10.1097/01.CCM.0000217919.22155.85
  112. Georgopoulos D., Matamis D., Routsi C., et al. Recombinant human erythropoietin therapy in critically ill patients: a dose-response study. Crit Care. 2005; 9(5): R508–R515. DOI: 10.1186/cc3786
  113. Zarychanski R., Turgeon A.F., McIntyre L., Fergusson D.A. Erythropoietin-receptor agonists in critically ill patients: a meta-analysis of randomized controlled trials. CMAJ. 2007; 177(7): 725–734. DOI: 10.1503/cmaj.071055
  114. Pammi M., Suresh G. Enteral lactoferrin supplementation for prevention of sepsis and necrotizing enterocolitis in preterm infants. Cochrane Database Syst. Rev. 2017; 6: CD007137. DOI: 10.1002/14651858.CD007137.pub5
  115. Lauterbach R., Kamińska E., Michalski P., Lauterbach J.P. Lactoferrin — a glycoprotein of great therapeutic potentials. Dev. Period. Med. 2016; 20(2): 118–125.
  116. Kruzel M.L., Zimecki M., Actor J.K. Lactoferrin in a Context of Inflammation-Induced Pathology. Front. Immunol. 2017; 8: 1438. DOI: 10.3389/fimmu.2017.01438
  117. Remy K.E., Cortés-Puch I., Solomon S.B., et al. Haptoglobin improves shock, lung injury, and survival in canine pneumonia. JCI Insight. 2018; 3(18). DOI: 10.1172/jci.insight.123013
  118. Kelly B.J., Lautenbach E., Nachamkin I., et al. Combined biomarkers discriminate a low likelihood of bacterial infection among surgical intensive care unit patients with suspected sepsis. Diagn. Microbiol. Infect. Dis. 2016; 85(1): 109–115. DOI: 10.1016/j.diagmicrobio.2016.01.003
  119. Immenschuh S., Vijayan V., Janciauskiene S., Gueler F. Heme as a Target for Therapeutic Interventions. Front. Pharmacol. 2017; 8: 146. DOI: 10.3389/fphar.2017.00146
  120. Thorburn T., Aali M., Kostek L., et al. Anti-inflammatory effects of a novel iron chelator, DIBI, in experimental sepsis. Clin. Hemorheol. Microcirc. 2017; 67(3–4): 241–250. DOI: 10.3233/CH-179205
  121. Ang M.T.C., Gumbau-Brisa R., Allan D.S., et al. A 3-hydroxypyridin-4-one chelator iron-binding polymer with enhanced antimicrobial activity. Medchemcomm. 2018; 9(7): 1206–1212. DOI: 10.1039/c8md00192h
Creative Commons License

This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License.