Anemia of chronic diseases: current state of the problem and perspectives

Cover Page

Cite item

Full Text

Abstract

Anemia of chronic diseases is a condition, that accompanies several chronic conditions, that have inflammation as an underlying cause. The article covers current concepts of pathogenesis, evaluation and treatment of this type of anemia. The new perspectives in the development of investigational methods and treatment are discussed. The new methods of iron deficiency assessment in patients with systemic inflammation are discussed separately.

About the authors

Olga I. Mironova

Sechenov First Moscow State Medical University (Sechenov University)

Author for correspondence.
Email: mironova_o_yu@staff.sechenov.ru
ORCID iD: 0000-0002-5820-1759

д-р мед. наук, проф. каф. факультетской терапии №1 Института клинической медицины им. Н.В. Склифосовского

Russian Federation, Moscow

Alexandr S. Panferov

Sechenov First Moscow State Medical University (Sechenov University)

Email: mironova_o_yu@staff.sechenov.ru

канд. мед. наук, доц. каф. факультетской терапии №1 Института клинической медицины им. Н.В. Склифосовского

Russian Federation, Moscow

References

  1. Matzner Y, Levy S, Grossowicz N, et al. Prevalence and Causes of Anemia in Elderly Hospitalized Patients. Gerontology. 1979;25(2):113-9. doi: 10.1159/000212328
  2. Weiss G. Pathogenesis and treatment of anaemia of chronic disease. Blood Rev. 2002;16(2):87-96. doi: 10.1054/blre.2002.0193
  3. Means Jr RT. Recent developments in the anemia of chronic disease. Curr Hematol Rep. 2003;2(2):116-21.
  4. World Health Organization. Noncommunicable diseases. Fact sheet. 2022. Available at: https://www.who.int/news-room/fact-sheets/detail/noncommunicable-diseases. Accessed: 02.06.2022.
  5. Bernell S, Howard SW. Use your words carefully: what is a chronic disease? Front Public Heal. 2016;4:159. doi: 10.3389/fpubh.2016.00159
  6. Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med. 2005;352(10):1011-23. doi: 10.1056/NEJMra041809
  7. Dulmovits BM, Tang Y, Papoin J, et al. HMGB1-Mediated Restriction of EPO Signaling Contributes to Anemia of Inflammation. Blood. 2022;139(21):3181-93. doi: 10.1182/blood.2021012048
  8. Yap GS, Stevenson MM. Inhibition of in vitro erythropoiesis by soluble mediators in Plasmodium chabaudi AS malaria: lack of a major role for interleukin 1, tumor necrosis factor alpha, and gamma interferon. Infect Immun. 1994;62(2):357-62.
  9. Gordeuk VR, Delanghe JR, Langlois MR, Boelaert JR. Iron status and the outcome of HIV infection: an overview. J Clin Virol. 2001;20(3):111-5. doi: 10.1016/s1386-6532(00)00134-7
  10. Fernández-García V, González-Ramos S, Avendaño-Ortiz J, et al. NOD1 splenic activation confers ferroptosis protection and reduces macrophage recruitment under pro-atherogenic conditions. Biomed Pharmacother. 2022;148:112769. doi: 10.1016/j.biopha.2022.112769
  11. Kautz L, Jung G, Valore EV, et al. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat Genet. 2014;46(7):678-84. doi: 10.1038/ng.2996
  12. De Domenico I, Ward DM, Langelier C, et al. The molecular mechanism of hepcidin-mediated ferroportin down-regulation. Mol Biol Cell. 2007;18(7):2569-78. doi: 10.1091/mbc.e07-01-0060
  13. Jiang L, Wang J, Wang K, et al. RNF217 regulates iron homeostasis through its E3 ubiquitin ligase activity by modulating ferroportin degradation. Blood. 2021;138(8):689-705. doi: 10.1182/blood.2020008986
  14. Diepeveen LE, Stegemann G, Wiegerinck ET, et al. Investigating the Molecular Mechanisms of Renal Hepcidin Induction and Protection upon Hemoglobin-Induced Acute Kidney Injury. Int J Mol Sci. 2022;23(3):1352. doi: 10.3390/ijms23031352
  15. Wieczorek M, Schwarz F, Sadlon A, et al. Iron deficiency and biomarkers of inflammation: A 3-year prospective analysis of the DO-HEALTH trial. Aging Clin Exp Res. 2022;34(3):515-25. doi: 10.1007/s40520-021-01955-3
  16. Yağcı S, Serin E, Acicbe Ö, et al. The relationship between serum erythropoietin, hepcidin, and haptoglobin levels with disease severity and other biochemical values in patients with COVID-19. Int J Lab Hematol. 2021;43:142-51. doi: 10.1111/ijlh.13479
  17. Zhou C, Chen Y, Ji Y, et al. Increased serum levels of hepcidin and ferritin are associated with severity of COVID-19. Med Sci Monit Int Med J Exp Clin Res. 2020;26:e926178-1. doi: 10.12659/MSM.926178
  18. Zuin M, Rigatelli G, Quadretti L, et al. Prognostic Role of Anemia in COVID-19 Patients: A Meta-Analysis. Infect Dis Rep. 2021;13(4):930-7. doi: 10.3390/idr13040085
  19. Chen Y, Xu W, Yang H, et al. Serum Levels of Hepcidin in Rheumatoid Arthritis and Its Correlation with Disease Activity and Anemia: A Meta-analysis. Immunol Invest. 2021;50(2-3):243-58. doi: 10.1080/08820139.2020.1742731
  20. Szymczak A, Zalewska M, Rymer W, Jankowska EA. Asymptomatic Human Immunodeficiency Virus-1 Infection with High CD4+ T Cell Count Does Not Alter Iron Metabolism or Hepcidin Levels: The Pilot Study. Infect Dis Ther. 2022;11(1):265-75. doi: 10.1007/s40121-021-00560-1
  21. Blindar VN, Dobrovolskaya MM, Khagazheeva MN, et al. The role of interleukin-6 and hepcidin 25 in the pathogenesis of anemic syndrome associated with malignant neoplasms with breast cancer patients before neoadjuvant chemotherapy. Klin Lab Diagn. 2021;66(3):147-53. doi: 10.51620/0869-2084-2021-66-3-147-153
  22. Han J, Wang K. Clinical significance of serum hepcidin in the diagnosis and treatment of patients with anemia of chronic disease: a meta-analysis. Biomarkers. 2021;26(4):296-301. doi: 10.1080/1354750X.2021.1893812
  23. Renders L, Budde K, Rosenberger C, et al. First-in-human Phase I studies of PRS-080# 22, a hepcidin antagonist, in healthy volunteers and patients with chronic kidney disease undergoing hemodialysis. PLoS One. 2019;14(3):e0212023. doi: 10.1371/journal.pone.0212023
  24. Boyce M, Warrington S, Cortezi B, et al. Safety, pharmacokinetics and pharmacodynamics of the anti-hepcidin Spiegelmer lexaptepid pegol in healthy subjects. Br J Pharmacol. 2016;173(10):1580-8. doi: 10.1111/bph.13433
  25. van Eijk LT, John ASE, Schwoebel F, et al. Effect of the antihepcidin Spiegelmer lexaptepid on inflammation-induced decrease in serum iron in humans. Blood. 2014;124(17):2643-6. doi: 10.1182/blood-2014-03-559484
  26. Macdougall IC, Rumjon A, Cinco J, et al. FP660 Pharmacokinetics and Pharmacodynamics of Lexaptepid, a Novel Anti-Hepcidin Molecule, in ESA-Resistant Haemodialysis Patients. Nephrol Dial Transplanation. 2015;30(suppl_3):iii294-5.
  27. Vadhan-Raj S, Abonour R, Goldman JW, et al. A first-in-human phase 1 study of a hepcidin monoclonal antibody, LY2787106, in cancer-associated anemia. J Hematol Oncol. 2017;10(1):1-12. doi: 10.1186/s13045-017-0427-x
  28. Sakamoto S, Kirinashizawa M, Mohara Y, Watanabe Y. Generation and characterization of monoclonal antibodies against mature hepcidin and its application to neutralization and quantitative alteration assay. Biosci Biotechnol Biochem. 2021;85(2):340-50. doi: 10.1093/bbb/zbaa013
  29. Than MM, Koonyosying P, Ruangsuriya J, et al. Effect of Recombinant Human Erythroferrone Protein on Hepcidin Gene (Hamp1) Expression in HepG2 and HuH7 Cells. Materials (Basel). 2021;14(21):6480. doi: 10.3390/ma14216480
  30. Lugus JJ, Park C, Ma YD, Choi K. Both primitive and definitive blood cells are derived from Flk-1+ mesoderm. Blood. 2009;113(3):563-6. doi: 10.1182/blood-2008-06-162750
  31. Sidiartha IGL, Bakta IM, Wiryana IM, et al. Eicosapentaenoic acid and docosahexaenoic acid in fish oil capsule supplementation in obese children decreases serum interleukin-6 and hepcidin and improves iron status. Bali Med J. 2017;6(1):97-101.
  32. Lainé F, Laviolle B, Bardou-Jacquet E, et al. Curcuma decreases serum hepcidin levels in healthy volunteers: a placebo-controlled, randomized, double-blind, cross-over study. Fundam Clin Pharmacol. 2017;31(5):567-73. doi: 10.1111/fcp.12288
  33. Barrington P, Sheetz MJ, Callies S, et al. Safety, tolerability, pharmacokinetics and pharmacodynamics of an anti-ferroportin antibody in patients with anemia due to chronic renal failure. Blood. 2016;128(22):1280.
  34. Isaacs JD, Harari O, Kobold U, et al. Effect of tocilizumab on haematological markers implicates interleukin-6 signalling in the anaemia of rheumatoid arthritis. Arthritis Res Ther. 2013;15(6):1-6. doi: 10.1186/ar4397
  35. Song SNJ, Iwahashi M, Tomosugi N, et al. Comparative evaluation of the effects of treatment with tocilizumab and TNF-α inhibitors on serum hepcidin, anemia response and disease activity in rheumatoid arthritis patients. Arthritis Res Ther. 2013;15(5):1-10. doi: 10.1186/ar4323
  36. Loza M, Campbell K, Sweet K, et al. FRI0243 Sirukumab treatment reduces levels of iron-regulatory proteins and ameliorates inflammation-associated anemia in rheumatoid arthritis patients. BMJ Publishing Group Ltd, 2017.
  37. Vallurupalli M, MacFadyen JG, Glynn RJ, et al. Effects of interleukin-1β inhibition on incident anemia: exploratory analyses from a randomized trial. Ann Intern Med. 2020;172(8):523-32. doi: 10.7326/M19-2945
  38. Scarozza P, De Cristofaro E, Scucchi L, et al. Effect of Vedolizumab on Anemia of Chronic Disease in Patients with Inflammatory Bowel Diseases. J Clin Med. 2020;9(7):2126. doi: 10.3390/jcm9072126
  39. Ghanim H, Abuaysheh S, Hejna J, et al. Dapagliflozin suppresses hepcidin and increases erythropoiesis. J Clin Endocrinol Metab. 2020;105(4):e1056-63. doi: 10.1210/clinem/dgaa057
  40. Zheng L, Tian J, Liu D, et al. Efficacy and safety of roxadustat for anaemia in dialysis-dependent and non-dialysis-dependent chronic kidney disease patients: A systematic review and meta-analysis. Br J Clin Pharmacol. 2022;88(3):919-32. doi: 10.1111/bcp.15055
  41. Xiong L, Zhang H, Guo Y, et al. Efficacy and Safety of Vadadustat for Anemia in Patients With Chronic Kidney Disease: A Systematic Review and Meta-Analysis. Front Pharmacol. 2021;12:795214. doi: 10.3389/fphar.2021.795214
  42. Bohlius J, Bohlke K, Castelli R, et al. Management of cancer-associated anemia with erythropoiesis-stimulating agents: ASCO/ASH clinical practice guideline update. Blood Adv. 2019;3(8):1197-210. doi: 10.1182/bloodadvances.2018030387
  43. Анемия при злокачественных новообразованиях. Клинические рекомендации Минздрава РФ. М., 2022 [Anemiia pri zlokachestvennykh novoobrazovaniiakh. Klinicheskiie rekomendatsii Minzdrava RF. Moscow, 2022 (in Russian)].
  44. Neoh K, Page A, Chin-Yee N, et al. Practice review: Evidence-based and effective management of anaemia in palliative care patients. Palliat Med. 2022;02692163221081967. doi: 10.1177/02692163221081967
  45. Dignass AU, Gasche C, Bettenworth D, et al. European consensus on the diagnosis and management of iron deficiency and anaemia in inflammatory bowel diseases. J Crohn’s Colitis. 2015;9(3):211-22. doi: 10.1093/ecco-jcc/jju009
  46. Анемия при хронической болезни почек. Клинические рекомендации Минздрава РФ. 2022. Режим доступа: https://cr.minzdrav.gov.ru/schema/623_4. Ссылка активна на 02.06.2022 [Anemiia pri khronicheskoi bolezni pochek. Klinicheskie rekomendatsii Minzdrava RF. 2022. Available at: https://cr.minzdrav.gov.ru/schema/623_4. Accessed: 02.06.2022 (in Russian)].
  47. Sierpinski R, Josiak K, Suchocki T, et al. High soluble transferrin receptor in patients with heart failure: a measure of iron deficiency and a strong predictor of mortality. Eur J Heart Fail. 2021;23(6):919-32. doi: 10.1002/ejhf.2036
  48. Fitzsimons S, Poppe KK, Choi Y, et al. Relationship between soluble transferrin receptor and clinical outcomes in patients with Heart Failure According to Ejection Fraction Phenotype: The New Zealand PEOPLE Study. J Card Fail. 2022;28(8):1255-63. doi: 10.1016/j.cardfail.2021.12.018
  49. Punnonen K, Irjala K, Rajamäki A. Serum transferrin receptor and its ratio to serum ferritin in the diagnosis of iron deficiency. Blood. 1997;89(3):1052-7.
  50. Fletcher A, Forbes A, Svenson N, et al. Guideline for the laboratory diagnosis of iron deficiency in adults (excluding pregnancy) and children. Br J Haematol. 2021;196(3):523-9. doi: 10.1111/bjh.17900
  51. Sullivan PS, Hanson DL, Chu SY, et al. Epidemiology of anemia in human immunodeficiency virus (HIV)-infected persons: results from the multistate adult and adolescent spectrum of HIV disease surveillance project. Blood. 1998;91(1):301-8.
  52. Nissenson AR, Goodnough LT, Dubois RW. Anemia: not just an innocent bystander? Arch Intern Med. 2003;163(12):1400-4. doi: 10.1001/archinte.163.12.1400
  53. Harrison L, Shasha D, Shiaova L, et al. Prevalence of anemia in cancer patients undergoing radiation therapy. Semin Oncol. 2001:54-9.
  54. Ludwig H, Fritz E, Leitgeb C, et al. Prediction of response to erythropoietin treatment in chronic anemia of cancer. Blood. 1994;84(4):1056-63.
  55. Rizzo JD, Lichtin AE, Woolf SH, et al. Use of epoetin in patients with cancer: evidence-based clinical practice guidelines of the American Society of Clinical Oncology and the American Society of Hematology. Blood. 2002;100(7):2303-20. doi: 10.1182/blood-2002-06-1767
  56. Gasche C, Waldhoer T, Feichtenschlager T, et al. Prediction of response to iron sucrose in inflammatory bowel disease-associated anemia. Am J Gastroenterol. 2001;96(8):2382-7. doi: 10.1111/j.1572-0241.2001.04094.x
  57. Wilson A, Reyes E, Ofman J. Prevalence and outcomes of anemia in inflammatory bowel disease: a systematic review of the literature. Am J Med. 2004;116(7):44-9. doi: 10.1016/j.amjmed.2003.12.011
  58. Müller HM, Horina JH, Kniepeiss D, et al. Characteristics and clinical relevance of chronic anemia in adult heart transplant recipients. Clin Transplant. 2001;15(5):343-8. doi: 10.1034/j.1399-0012.2001.150507.x
  59. Maheshwari A, Mishra R, Thuluvath PJ. Post-liver-transplant anemia: Etiology and management. Liver Transplant. 2004;10(2):165-73. doi: 10.1002/lt.20031
  60. Collins AJ, Li S, Peter WST, et al. Death, hospitalization, and economic associations among incident hemodialysis patients with hematocrit values of 36 to 39%. J Am Soc Nephrol. 2001;12(11):2465-73. doi: 10.1681/ASN.V12112465
  61. Locatelli F, Pisoni RL, Combe C, et al. Anaemia in haemodialysis patients of five European countries: association with morbidity and mortality in the Dialysis Outcomes and Practice Patterns Study (DOPPS). Nephrol Dial Transplant. 2004;19(1):121-32. doi: 10.1093/ndt/gfg458
  62. Stenvinkel P. The role of inflammation in the anaemia of end-stage renal disease. Nephrol Dial Transplant. 2001;16(suppl_7):36-40. doi: 10.1093/ndt/16.suppl_7.36
  63. Thomas C, Thomas L. Biochemical markers and hematologic indices in the diagnosis of functional iron deficiency. Clin Chem. 2002;48(7):1066-76
  64. Ermens AA, Hoffmann JJ, Krockenberger M, Van Wijk EM. New erythrocyte and reticulocyte parameters on CELL-DYN Sapphire: analytical and preanalytical aspects. Int J Lab Hematol. 2012;34(3):274-82. doi: 10.1111/j.1751-553X.2011.01391.x
  65. Urrechaga E, Borque L, Escanero JF. Percentage of hypochromic erythrocytes as a potential marker of iron availability. Clin Chem Lab Med. 2011;50(4):685-7. doi: 10.1515/cclm.2011.837
  66. Urrechaga E, Unceta M, Borque L, Escanero JF. Low hemoglobin density potential marker of iron availability. Int J Lab Hematol. 2012;34(1):47-51. doi: 10.1111/j.1751-553X.2011.01355.x
  67. Brugnara C, Schiller B, Moran J. Reticulocyte hemoglobin equivalent (Ret He) and assessment of iron-deficient states. Clin Lab Haematol. 2006;28(5):303-8. doi: 10.1111/j.1365-2257.2006.00812.x
  68. Buttarello M, Rauli A, Mezzapelle G. Reticulocyte count and extended reticulocyte parameters by Mindray BC-6800: Reference intervals and comparison with Sysmex XE-5000. Int J Lab Hematol. 2017;39(6):596-603. doi: 10.1111/ijlh.12705

Copyright (c) 2022 Consilium Medicum

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


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies