Acessibilidade / Reportar erro

Resistance of dialyzed patients to erythropoietin

Abstract

Resistance to recombinant human erythropoietin is a common condition in dialyzed patients with chronic kidney disease and is associated with more hospitalizations, increased mortality and frequent blood transfusions. The main cause of hyporesponsiveness to recombinant human erythropoietin in these patients is iron deficiency. However, a high proportion of patients does not respond to treatment, even to the use of intravenous iron, which indicates the presence of other important causes of resistance. In addition to the iron deficiency, the most common causes of resistance include inflammation, infection, malnutrition, inadequate dialysis, and hyperparathyroidism, although other factors may be associated. In the presence of adequate iron stores, other causes should be investigated and treated appropriately.

Dialysis; Erythropoietin; Iron


Introduction

Chronic kidney disease (CKD) is considered a public health problem worldwide with high incidence and prevalence rates.1 Romão Junior JE. A doença renal crônica: do diagnóstico ao tratamento. Prática Hospitalar. 2007;52:183-7.In end-stage renal disease (ESRD), renal function must be replaced by dialysis or renal transplantation.2Manual de Transplante Renal. Associação Brasileira de Transplantes de Órgãos (ABTO). Available from: http://www.abto.org.br/abtov03/Upload/file/ProfissionalManual/manualtransplanterim.pdf [cited 31.10.14].
http://www.abto.org.br/abtov03/Upload/fi...
In Brazil, the number of patients on dialysis has increased gradually over the years. According to the Sociedade Brasileira de Nefrologia (SBN), 42,695 and 100,397 patients were under dialysis in 2000 and 2013, respectively.3Censo da Sociedade Brasileira de Nefrologia 2013. São Paulo: Sociedade Brasileira de Nefrologia (SBN). Available from: http://www.sbn.org.br/pdf/censo2013-14-05.pdf [cited 31.10.14].
http://www.sbn.org.br/pdf/censo2013-14-0...

Anemia is one of the most frequent early complications of CKD.4Kazmi WH, Kausz AT, Khan S, Abichandani R, Ruthazer R, Obrador GT, et al. Anemia: an early complication of chronic renal insufficiency. Am J Kidney Dis. 2001;38(4):803-12. The main cause is erythropoietin (EPO) deficiency due to impaired kidney function. However, other causes should be considered when the severity of anemia is inconsistent with the decrease in renal function; when there is evidence of iron deficiency or matching decreases in hemoglobin, leukopenia and/or thrombocytopenia are also found.5National Kidney Foundation. K/DOQI clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney disease. Am J Kidney Dis.2006;26 Suppl. 3:1-145.

The treatment of anemia in CKD patients usually involves the use of recombinant human erythropoietin (rHuEPO). The main cause of rHuEPO treatment failure is the loss or low iron availability.6Macdougall IC, Hutton RD, Coles GA, William JD. The use of erythropoietin in renal failure. Postgrad Med J. 1991;67(783):9-15. The prevalence of iron deficiency is very common in CKD, affecting as many as 50% of patients.7Hutchinson FN, Jones WJ. A cost-effectiveness analysis of anemia screening before erythropoietin in patients with endstage renal disease. Am J Kidney Dis.1997;29(5):651-7. However, despite rHuEPO and intravenous iron in the majority of patients, the prevalence of anemia reaches 34% in Brazil.8Sesso RC, Lopes AA, Thome FS, Lugon JR, Watanabe Y, Santos DR. Relatório do Censo Brasileiro de Diálise 2012. J Bras Nefrol. 2014;36(1):48-53. This indicates the existence of other important factors related to rHuEPO resistance.

The definition of anemia in CKD patients has changed with some guidelines being produced over the last few years. In 2004 the Revised European Best Practice Guidelines (EBPG) on Anemia defined low hemoglobin levels as values <11.5 g/dL in adult females and <13.5 g/dL in adult males (<12 g/dL in over 70-year olds). Patients with CKD should maintain a hemoglobin level >11 g/dL (hematocrit >33%). In addition, levels >12 g/dL are not recommended for patients with severe cardiovascular disease.9Locatelli F, Aljama P, Barany P, Canaud B, Carrera F, Eckardt KU, et al. Revised European best practice guidelines for the management of anaemia in patients with chronic renal failure. Nephrol Dial Transplant. 2004;19 Suppl. 2:ii1-47.

An update of the 2006 National Kidney Foundation Kidney Disease Outcomes Quality Initiative (NKF/KDOQI) guidelines in 2007 suggested that anemia is associated with hemoglobin levels <13.5 g/dL in adult males and <12.0 g/dL in adult females. In patients with CKD, hemoglobin should be between 11 and 12 g/dL, however hemoglobin targets greater than 13 g/dL may increase the risk for serious adverse effects and are not recommended.5National Kidney Foundation. K/DOQI clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney disease. Am J Kidney Dis.2006;26 Suppl. 3:1-145. , 1010 KDOQI. KDOQI Clinical Practice Guideline and Clinical Practice Recommendations for anemia in chronic kidney disease: 2007 update of hemoglobin target. Am J Kidney Dis.2007;50(3):471-530.

The KDOQI modified the EBPG definition defining anemia in adult males as hemoglobin <13.5 g/dL regardless of age since the decrease in hemoglobin levels among over 60-year-old males is frequently related to concurrent diseases. In addition, in adult females the hemoglobin target is 12 g/dL. The European Renal Best Practice (ERBP) Work Group agrees with the KDOQI definitions.1111 Locatelli F, Covic A, Eckardt KU, Wiecek A, Vanholder R, ERA-EDTA ERBP Advisory Board. Anaemia management in patients with chronic kidney disease: a position statement by the Anaemia Working Group of European Renal Best Practice (ERBP). Nephrol Dial Transplant.2009;24(2):348-54.

Recently, the Kidney Disease: Improving Global Outcome (KDIGO) group defined anemia in adults and children aged >15 years with CKD when the hemoglobin levels are <13.0 g/dL in males and <12.0 g/dL in females.1212 KDIGO. KDIGO clinical practice guideline for anemia in chronic kidney disease. Kidney Int Suppl. 2012;2:279-335. Table 1 shows the definitions of anemia and hemoglobin targets in CKD patients.

Table 1
Definition of anemia and hemoglobin target in CKD patients.

Although there is no consensus about the definition for rHuEPO resistance, the evaluation of resistance is recommended if there is an increase ≥25% in erythropoietin dose or <1 mg/dL gain in hemoglobin levels after 2-4 weeks of treatment.1313., Locatelli F Pisoni RL, Combe C, Bommer J, Andreucci VE, Piera L, et al. Anaemia in hemodialysis 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.

According to the Brazilian Ministry of Health,1414 Brasil. Ministério da Saúde Protocolo Clínico e Diretrizes Terapêuticas. Anemia em pacientes com insuficiência renal crônica - alfaepoetina. Brasília: Ministério da Saúde; 2010. rHuEPO resistance is defined as a persistent anemia (hemoglobin <10-12 g/dL) or the necessity of very high erythropoietin doses of epoetin alfa (300 IU/kg/week subcutaneously or 450 IU/kg/week intravenously). Epoetin alfa should be initiated at a dose of 50-100 IU/kg subcutaneously, one to three times a week. The initial goal of treatment is to achieve a rate of weekly increase in hemoglobin levels of 0.3 g/dL. If after four weeks of treatment, this response is not observed and the hemoglobin remains below 11 g/dL, the dose should be increased by 25%. However, after four weeks if the hemoglobin level is greater than 13 g/dL, the drug should be suspended temporarily, since the maintenance of higher hemoglobin levels is associated with increased morbidity and mortality. The recommended therapeutic target is to preserve hemoglobin levels from 11 to 12 g/dL or hematocrit from 33% to 36%.5National Kidney Foundation. K/DOQI clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney disease. Am J Kidney Dis.2006;26 Suppl. 3:1-145.

Anemia in CKD is usually normocytic and normochromic. The characteristics of erythrocytes as determined by hematimetric indices, mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH) and mean corpuscular hemoglobin concentration (MCHC) can characterize the etiology of anemia.1515 Ribeiro-Alves MA, Gordan PA. Diagnóstico de Anemia em Pacientes Portadores de Doença Renal Crônica. J Bras Nefrol.2014;36 1 Suppl. 1:9-12. In addition to hematimetric indices, the laboratory investigation includes complete blood cell count, reticulocyte count, serum iron, determination of the transferrin saturation and serum ferritin, as well as occult blood in stools and the levels of folic acid and vitamin B12.1212 KDIGO. KDIGO clinical practice guideline for anemia in chronic kidney disease. Kidney Int Suppl. 2012;2:279-335. , 1616 Abensur H. Anemia da doenc¸a renal crônica. J Bras Nefrol.2004;26(3):26-8.

The biochemical markers of iron deficiency (serum iron, ferritin, transferrin saturation and soluble transferrin receptor - sTfR) have limited value in functional iron deficiency as they are changed in several clinical conditions such as the ones that evolve with rHuEPO therapy.1717 Brugnara C. Iron deficiency and erythropoiesis: new diagnostic approaches. Clin Chem. 2003;49(10):1573-8. However, reticulocyte hemoglobin content (CHr or Ret-He) is a sensitive indirect marker of iron deficiency, which reflects recent changes in erythropoiesis.1818 Mittman N, Sreedhara R, Mushnick R, Chattopadhyay J, Zelmanovic D, Vaseghi M, et al. Reticulocyte hemoglobin content predicts functional iron deficiency in hemodialysis patients receiving rHuEPO. Am J Kidney Dis. 1997;30(6):912-22. The measurement of CHr in peripheral blood samples is useful for assessing the amount of functional iron that was available in the bone marrow for new red blood cell production over the previous 3-4 days.1919 Mast AE, Blinder MA, Dietzen DJ. Reticulocyte hemoglobin content. Am J Hematol. 2008;83(4):307-10. CHr maybe a more sensitive marker of functional iron deficiency in patients receiving erythropoietin therapy.2020 Fishbane S, Shapiro W, Dutka P, Valenzuela OF, Faubert J. A randomized trial of iron deficiency testing strategies in hemodialysis patients. Kidney Int. 2001;60(6):2406-11. It may also be an early indicator of the effectiveness of iron replacement therapy.2121 Brugnara C, Laufer MR, Friedman AJ, Bridges K, Plat O. Reticulocyte hemoglobin content (CHr): early indicator of iron deficiency and response to therapy. Blood. 1994;83(10):3100-1.

Thomas and Thomas2222 Thomas C, Thomas L. Biochemical markers and hematologic indices in the diagnosis of functional iron deficiency. Clin Chem. 2002;48(7):1066-76. presented a novel approach to functional iron deficiency with the use of CHr and the percentage of hypochromic erythrocytes (HYPO). Functional iron deficiency was defined as a CHr <28 pg and a HYPO <5% based on the levels in healthy controls. Moreover, the sTfR-F index (sTfR/log ferritin), which reflects the iron store status, can be used to differentiate functional iron deficiency in states of iron depletion and iron repletion. A diagnostic strategy combined CHr and sTfR-F index to identify four major categories of iron deficiency: (1) iron repletion with normal erythropoiesis (CHr and sTfR-F index within appropriate reference values); (2) reduced iron supply with still-normal erythropoiesis (CHr within reference values; increased sTfR-F index); (3) depletion of stores and functional iron deficiency (reduced CHr; increased sTfR-F index); and (4) functional iron deficiency in a state of iron repletion (reduced CHr; sTfR-F index within reference values).

The strong association between anemia and cardiovascular complications should be highlighted. The decrease in tissue oxygenation causes tachycardia, vasodilation and increased cardiac work and may cause left ventricular hypertrophy.2323 Lea JP, Norris K, Agodoa L. The role of anemia management in improving outcomes for African Americans with chronic kidney disease. Am J Nephrol. 2008;28(5):732-43.

The development and persistence of anemia in patients with CKD are also associated with worse quality of life, reduced exercise capacity, decreased mental agility and renal function and increase the prevalence of hospitalization and mortality.1313., Locatelli F Pisoni RL, Combe C, Bommer J, Andreucci VE, Piera L, et al. Anaemia in hemodialysis 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.

The main causes of resistance to treatment with rHuEPO (Table 2) in dialysis patients are discussed in this review.

Table 2
Risk factors of resistance to recombinant human erythropoietin.

Causes of resistance to treatment with recombinant human erythropoietin in patients under dialysis

Iron deficiency

Iron deficiency or impairment of iron availability is the most frequent cause of rHuEPO treatment resistance in patients under dialysis.2424 Kalantar-zadeh K, Ikizler TA, Block G, Avram MM, Kopple JD. Malnutrition-inflammation complex syndrome in dialysis patients: causes and consequences. Am J Kidney Dis. 2003;42(5):864-81. , 2525 Bamgbola O. Resistance to erythropoietin-stimulating agents: etiology, evaluation, and therapeutic considerations. Pediatr Nephrol. 2010;27(2):195-205.

In these patients, the deficiency or reduction of total iron stores can occur due to an increase in demand of this nutrient during the production of red blood cells in the bone marrow. This absolute iron deficiency may also be related to thedialysis procedure, which promotes premature destruction of red blood cells (hemolysis), but also due to gastrointestinal bleeding, or frequent laboratory blood tests and surgeries, which patients can be submitted to.2626 Wish JB. Assessing iron status: beyond serum ferritin and transferrin saturation. Clin J Am Soc Nephrol. 2006; Suppl. 1:S4-8.

In functional iron deficiency, suitable stores of this nutrient can be observed, but the mobilization of iron to the blood-stream is insufficient to reach the demand of the erythroid marrow. This condition is common in inflammatory states due to the cytokines that block the release of iron from deposits.2727 Means RT, Krantz SB. Progress in understanding the pathogenesis of the anemia of chronic disease. Blood.1992;80(7):1639-47.

Iron deficiency anemia is characterized by microcytosis and hypochromia and a careful microscopic examination of erythrocytes may lead to suspicion of iron deficiency. Furthermore, transferrin saturation and serum ferritin levels may help to distinguish between conditions associated with defi- ciency or impairment in the availability of iron.5National Kidney Foundation. K/DOQI clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney disease. Am J Kidney Dis.2006;26 Suppl. 3:1-145.

Hepcidin, a small peptide synthesized mainly in hepatocytes, is the central regulator of systemic iron homeostasis.2828 Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B, Brissot P, et al. A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem. 2001;276(11):7811-9. , 2929 Rivera S, Nemeth E, Gabayan V, Lopez MA, Farshidi D, Ganz T. Synthetic hepcidin causes rapid dose-dependent hipoferremia and is concentrated in ferroportin-containing organs. Blood.2005;106(6):2196-9. Hepcidin binds to ferroportin, an iron transporter present on cells of the intestinal duodenum, macrophages, and cells of the placenta, and regulates iron release to the plasma. When hepcidin concentrations are low, molecules of ferroportin are exposed on the plasmatic membrane and release iron. When hepcidin levels increase, hepcidin binds to ferroportin inducing its internalization and degradation, thereby leading to reductions in iron release.3030 Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004;306(5704):2090-3. , 3131 De Domenico I, Ward DM, Kaplan J. Hepcidin regulation: ironing out the details. J Clin Invest. 2007;117(7): 1755-8.

Hepcidin concentration in turn is regulated by iron, erythropoietic activity and inflammation.3232., Nemeth E. Ganz T The role of hepcidin in iron metabolism. Acta Haematol. 2009;122(2-3):78-86. IL-6 induces hypoferremia during inflammation by inducing the synthesis of hepcidin caused by decreases in serum iron and transferrin saturation. In addition, this cytokine by itself rapidly induces hypoferremia. Since the process of erythropoiesis is the largest consumer of iron in the body, the decrease in iron supply reduces hemoglobin synthesis and can lead to anemia.3333. Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK, et al. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest.2004;113(9):1271-6.

Hepcidin deficiency or resistance to hepcidin is associated with iron overload in hereditary hemochromatosis, iron-loading anemia, and hepatitis C. Hepcidin excess or ferro-portin deficiency is the cause of iron-refractory iron deficiency anemia, ferroportin disease, anemia of inflammation, CKDs, and cancer.3434., Ganz T. Nemeth E Hepcidin and iron homeostasis. Biochim Biophys Acta. 2012;1823(9):1434-43.

Chronic inflammation and infection

The role of inflammation in the development of anemia in patients with CKD should be highlighted. It is known that the release of cytokines such as interferon-gamma (IFN- ), tumor necrosis factor alpha (TNF- ), interleukin 1 (IL-1) and interleukin 6 (IL-6) can induce rHuEPO erythroid progenitor cell resistance or impair the release of stored iron in the reticu- loendothelial system for the production of hemoglobin.35-37

Infectious diseases may also be related to anemia resulting in chronic inflammation.3838. Fuchs D, Hausen A, Reibnegger G, Werner ER, Werner-felmayer G, Dierich MP, et al. Immune activation and the anaemia associated with chronic inflammatory disorders. Eur J Haematol. 1991;46(2):65-70.

A relationship between cytomegalovirus (CMV) infection and lack of response to rHuEPO has been associated to increased production of proinflammatory cytokines such as IFN- and TNF- .3939. Betjes MG, Huisman M, Weimar W, Litjens NHR. Expansion of cytolytic CD4+ CD28− T cells in end-stage renal disease. Kidney Int.2008;74(6):760-7. , 4040. Cooper AC, Breen CP, Vyas B, Ochola J, Kemeny DM,. Macdougall IC Poor response to recombinant erythropoietin is associated with loss of T-lymphocyte CD28 expression and altered interleukin-10 production. Nephrol Dial Transplant.2003;18(1):133-40. Betjes et al.4141. Betjes MG, Weimar W,. Litjens NHR CMV seropositivity determines epoetin dose and hemoglobin levels in patients with CKD. J Am Soc Nephrol. 2009;20(12):2661-6. showed an association between CMV and low hemoglobin levels in patients with CKD undergoing hemodialysis and high doses of rHuEPO.

Besides chronic inflammatory process, human parvovirus B19 (B19) can also cause anemia due to infection and lysis of erythroid precursors in the bone marrow.4242. Brown KE, Anderson SM, Young NS. Erythrocyte P antigen: cellular receptor for B19 parvovirus. Science.1993;262(5130):114-7. , 4343. Chisaka H, Morita E, Yaegashi N, Sugamura K. Parvovirus B19 and the pathogenesis of anaemia. Rev Med Virol. 2003;13(6):347-59. This infection may worsen in patients who have increased red blood cell destruction, as observed in patients undergoing dialysis treatment. Moreover, these patients are under increased risk of contracting HIV infection through blood transfusion or hemodialysis.44-47

Although some cases of B19 infection in patients undergoing dialysis have been reported, there are few studies which evaluated the incidence and clinical significance of B19 infection in these patients.48-50 Generally, this viral infection has been associated with transient aplastic crisis and unresponsiveness to treatment with rHuEPO.4646. Ozeki M, Fukushima T, Ohzeki M, Sasaki T, Kashihara N. A nosocomial parvovirus B19 infection-induced transient aplastic crisis in a patient with chronic renal failure. Clin Nephrol. 2006;65(2):141-3. , 5151. Duranay M, Bali M, Sahin M, Yakinci G, Vurgun N, Dilmen U. Parvovirus B19 infection and unresponsiveness to erythropoietin therapy in haemodialysis patients. Nephrol Dial Transplant.1998;13(3):779-80. However, several studies have reported the occurrence of acquired pure red cell aplasia (PRCA) and severe transfusion-dependent anemia in kidney transplant patients infected by B19.4848. Becker MR, Schneider B, Reber U, Poge U, Klein B, Klehr HU, et al. Renal anemia aggravated by long-term parvovirus B19 and cytomegalovirus infection in a renal transplant patient: case report and evaluation of B19 seroprevalence in dialysis patients. Transplant Proc. 2005;37(10):4306-8. , 52-55 PRCA is a disorder characterized by anemia that leads to the almost complete absence of erythroid cells from precursors in the bone marrow but with normal production of granulocytes and megakaryocytes. Besides B19 infection, which has tropism for erythroid precursors in the bone marrow, other conditions that result in PRCA include the presence of autoantibodies directed against red lineage progenitors, transient erythroblastopenia in childhood, pregnancy, leukemia, infectious processes, toxins, and the use of some drugs.5656. Fish P, Handgretinger R, Shaefer H. Pure red cell aplasia. Br J Haematol. 2000;111(4):1010-22.

Cofactor deficiency and malnutrition

In patients under dialysis, the main causes of protein energy malnutrition include low intake of nutrients; muscle loss due to increased protein catabolism and decrease in their synthesis; insulin resistance; loss of nutrients by dialysis and oxidative stress.57 The inflammatory process is also a major cause of protein energy malnutrition, which may occur in 13-51% of patients under hemodialysis.5858. Qureshi AR, Alvestrand A, Danielsson A, Divino-Filho JC, Gutierrez A, Lindholm B, et al. Factors predicting malnutrition in hemodialysis patients: a cross-sectional study. Kidney Int.1998;53(3):773-82.

Malnutrition has been associated with resistance to treatment with rHuEPO in patients under dialysis. Laboratory tests show low percentages of transferrin saturation index, low serum albumin concentrations and body mass index (BMI), but high levels of C-reactive protein (CRP) in these patients.5959., Locatelli F Andrulli S, Memoli B, Maffei C, Vecchio LD, Aterini S, et al. Nutritional-inflammation status and resistance to erythropoietin therapy in haemodialysis patients. Nephrol Dial Transplant.2006;21:991-8. , 6060. Kalantar-Zadeh K, Lee GH, Miller JE, Streja E, Jing J, Robertson JA, et al. Predictors of hyporesponsiveness to erythropoiesis-stimulating agents in hemodialysis patients. Am J Kidney Dis.2009;53(5):823-34. In addition to be a marker of iron stores, ferritin may also be increased in malnutrition.6161. Gaweda AE, Goldsmith LJ, Brier ME, Aronoff GR. Iron, inflammation, dialysis adequacy, nutritional status, and hyperparathyroidism modify erythropoietic response. Clin J Am Soc Nephrol. 2010;5(4):576-81.

Deficiencies of folic acid and vitamin B12 may be associ- ated with anemia and resistance to treatment with rHuEPO. Thus, when macrocytosis is detected, the levels of these nutrients should be evaluated.6262. Shaefer R, Teschner M, Kosch M. Folate metabolism in renal failure. Nephrol Dial Transplant.2002;17(S5):24-7. Besides the changes in erythropoiesis, folic acid and vitamin B12 deficiencies can lead to increases in homocysteine levels, which in turn is associated with an increased risk of cardiovascular complications in renal patients.6363. Perna AF, Ingrosso D, Santo NG, Galletti P, Brunone M, Zappia V. Metabolic consequences of folate-induced reduction of hyperhomocysteinemia in uremia. Am Soc Nephrol. 1997;8(12):1899-905. , 6464. Vecchi AF, Bamonti-Catena F, Finazzi S, Campolo J, Taioli E, Novembrino C, et al. Homocysteine, vitamin b12, and serum and erythrocyte folate in peritoneal dialysis and hemodialysis patients. Perit Dial Int. 2000;20(2):169-73.

Inadequate dialysis

In patients with CKD, damage to erythrocytes can occur in the presence of uremic toxins, which also inhibit the production of EPO and erythropoiesis. Furthermore, the dialysis procedure causes mechanical damage to erythrocytes, and leads to blood loss.6565. Costa E, Belo L, Santos-Silva A. Special problems in hemodialysis patients Internet. In: Resistance to recombinant human erythropoietin therapy in haemodialysis patients. Portugal: In Tech; 2011 [Chapter 3]. Available from: http://www.intechopen.com/books/special-problems-in-hemodialysis-patients/resistance-to-recombinant-human-erythropoietin-therapy-in-haemodialysis-patients [cited 14.10.13].
http://www.intechopen.com/books/special-...
, 6666. Shahab I, Khanna R, Nolph KD. Peritoneal dialysis or hemodialysis? A dilemma for the nephrologist. Adv Perit Dial. 2006;22:180-5.

The inadequacy of the dialysis dose is an important cause of anemia in patients under dialysis. In order to evaluate whether the dialysis procedure is removing enough uremic toxins, the patient's blood is sampled at the start and at the end of dialysis. The levels of urea in the two blood samples are then compared. In the Kt/V method, the dialyzer urea clearance (K) is multiplied by dialysis time (t), and the product divided by the patient's urea distribution volume (V). According to the NKF/KDOQI guidelines for patients under hemodialysis, the Kt/V target is ≥1.3, and in patients under peritoneal dialysis the target is ≥1.7/week.6767. National Kidney Foundation. K/DOQI clinical practice guidelines for nutrition in chronic renal failure. Am J Kidney Dis.2000;35 6 Suppl. 2:S1-40. A study by Gaweda et al.6161. Gaweda AE, Goldsmith LJ, Brier ME, Aronoff GR. Iron, inflammation, dialysis adequacy, nutritional status, and hyperparathyroidism modify erythropoietic response. Clin J Am Soc Nephrol. 2010;5(4):576-81. showed that patients with adequate dialysis assessed by Kt/V, require lower doses of rHuEPO. Although the pathophysiological mechanism that links inadequate dialysis to the lack of response to rHuEPO is still not completely understood, other factors such as inflammation and vascular access complications may be associated with poorer response to treatment.

The adequacy of the dose of dialysis is also related to a decrease in costs, since patients with the best values of Kt/V require smaller doses of rHuEPO.6868. Movilli E, Cancarini GC, Zani R, Camerini C, Sandrini M, Maiorca R. Adequacy of dialysis reduces the doses of recombinant erythropoietin independently from the use of biocompatible membranes in haemodialysis patients. Nephrol Dial Transplant.2001;16(1):111-4.

Hyperparathyroidism

Hyperparathyroidism, characterized by increased parathyroid hormone (PTH), is associated with lack of response to treatment with rHuEPO due to endogenous EPO inhibition, reduction of erythroid precursors in the bone marrow and erythrocyte survival. This hormone is also associated to the induction of bone marrow fibrosis.6060. Kalantar-Zadeh K, Lee GH, Miller JE, Streja E, Jing J, Robertson JA, et al. Predictors of hyporesponsiveness to erythropoiesis-stimulating agents in hemodialysis patients. Am J Kidney Dis.2009;53(5):823-34. , 6969. Druek TB, Eckard K. Role of secondary hyperparathyroidism in erythropoietin resistance of chronic renal failure patients. Nephrol Dial Transplant.2002;17(S5):28-31. , 7070. Brancaccio D, Cozzolino M, Gallieni M. Hyperparathyroidism and anemia in uremic subjects: a combined therapeutic approach. J Am Soc Nephrol. 2004;15 Suppl. 1:S21-4.

According to the NKF/KDOQI,7171. National Kidney Foundation. Kidney disease-dialysis outcome quality initiative: K/DOQI Clinical Practice Guidelines for bone metabolism and disease in chronic kidney disease. Am J Kidney Dis.2003;42 4 Suppl. 3:S1-202. PTH levels between 150 and 300 pg/mL are desirable in patients undergoing dialysis. However, the threshold at which PTH levels could affect the response to rHuEPO remains unclear. Rao et al.7272. Rao DS, Shih M, Mohini R. Effect of serum parathyroid hormone and bone marrow fibrosis on the response to erythropoietin in uremia. N Engl J Med.1993;328(3):171-5. demonstrated that patients who responded to treatment with rHuEPO had lower PTH levels (around 266 ± 322 pg/mL) compared with those who did not respond to treatment, with mean levels of 800 ± 248 pg/mL. Another study by Gaweda et al.6161. Gaweda AE, Goldsmith LJ, Brier ME, Aronoff GR. Iron, inflammation, dialysis adequacy, nutritional status, and hyperparathyroidism modify erythropoietic response. Clin J Am Soc Nephrol. 2010;5(4):576-81. demonstrated that PTH levels of 300, 600 and 900 pg/mL were associated with approximately 90%, 79% and 67% of the maximum response to treatment with rHuEPO, respectively.

Angiotensin-converting enzyme inhibitors and angiotensin II type 1 receptor blockers

The renin-angiotensin system was previously only thought to affect the cardiovascular system. However, this system plays also an important role in hematopoiesis which explains the reduction in hematocrit levels or anemia as a side effect of treatment using angiotensin-converting enzyme inhibitors (ACE inhibitors) and angiotensin II type 1 receptor blockers (ARBs).7373. Vlahakos DV, Marathias KP, Madias NE. The role of the renin-angiotensin system in the regulation of erythropoiesis. Am J Kidney Dis.2010;56(3):558-65. , 7474. Rodgers KE, diZerega JS. Contribution of the local RAS to hematopoietic function: a novel therapeutic target. Front Endocrinol (Lausanne). 2013;23(4):157.

The ACE, which plays a central role in blood pressure control system,7575. Erdös EG. Angiotensin I converting enzyme and the changes in our concepts through the years. Hypertension. 1990;16(4):363-70. is also responsible for the hydrolysis of acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), a tetrapeptide which naturally occurs in many body tissues. The physiological AcSDKP is a negative regulator of erythropoiesis that inhibits the entry of hematopoietic stem cells in the S phase of the cell cycle, keeping them in phase G0.7676. Lenfant M, Wdzieczak-bakala J, Guittet E, Prome JC, Sotty D, Frindel E. Inhibitor of hematopoietic pluripotent stem cell proliferation: purification and determination of its structure. Proc Natl Acad Sci USA. 1989;86(3):779-82. , 7777. Rieger K, Saez-servent N, Papet MP, Wdzieczak-Bakala J, Morgat J, Thierry J, et al. Involvement of human plasma angiotensin I-converting enzyme in the degradation of the haemoregulatory peptide N-acetyl seryl aspartyl lysyl proline. Biorem J. 1993;296(Pt. 2):373-8. Studies have shown that the use of ACE inhibitors is associated with increased plasma concentrations of this tetrapeptide. Thus, patients taking antihypertensive ACE inhibitors may be resistant to treatment with rHuEPO.7878. Azizi M, Rousseau A, Ezan E, Guyene T, Michelet S, Grognet J, et al. Acute angiotensin-converting enzyme inhibition increases the plasma level of the natural stem cell regulator N-acetyl-seryl-aspartyl-lysyl-proline. J Clin Invest. 1991;97(3):839-44. , 7979. Meur YL, Lorgeot V, Comte L, Szelaq JC, Leroux-Robert C, Praloran V. Plasma levels and metabolism of AcSDKP in patients with chronic renal failure: relationship with erythropoietin requirements. Am J Kidney Dis.2001;38(3):510-7. The lack of angiotensin II production, due to an interruption of the renin-angiotensin system, is a direct cause of anemia, indicating that angiotensin II regulates hematopoiesis.8080. Cole J, Ertoy D, Lin H, Sutliff RL, Ezan E, Guyene TT, et al. Lack of angiotensin II - facilitated erythropoiesis causes anemia in angiotensin-converting enzyme-deficient mice. J Clin Invest.2000;106(11):1391-8. Angiotensin II acts as a growth factor and directly stimulates proliferation of erythroid progenitors in the bone marrow. Additionally, angiotensin II enhances EPO secretion, which results in increased red blood cell mass.7373. Vlahakos DV, Marathias KP, Madias NE. The role of the renin-angiotensin system in the regulation of erythropoiesis. Am J Kidney Dis.2010;56(3):558-65.

Decreases in hemoglobin levels occur in adults with CKD after therapy with ACE inhibitors and/or ARBs.8181. Iodice C, Balletta MM, Minutolo R, Giannattasio P, Tuccillo S, Bellizzi V, et al. Maximal suppression of renin-angiotensin system in nonproliferative glomerulonephritis. Kidney Int.2003;63(6):2214-21. , 8282. Jacobsen P, Andersen S, Jensen BR, Parving HH. Additive effect of ACE inhibition and angiotensin II receptor blockade in type I diabetic patients with diabetic nephropathy. J Am Soc Nephrol. 2003;14(4):992-9. These drugs have been associated with a dose-dependent decrease in hematocrit and anemia and should be considered in the differential diagnosis of anemia in patients with a variety of illnesses including renal transplantation, decreased kidney function and heart failure. Since this effect can be reversible, the decision to decrease the dose or discontinue ACE inhibitors or ARBs therapy should consider the severity of the clinical condition and availability of alternative therapies.8383. Marathias KP, Agroyannis B, Mavromoustakos T, Matsoukas J,. Vlahakos DV Hematocrit-lowering effect following inactivation of renin-angiotensin system with angiotensin converting enzyme inhibitors and angiotensin receptor blockers. Curr Top Med Chem. 2004;4(4):483-6.

Anti-erythropoietin antibodies

Although treatment with rHuEPO is well tolerated by most patients, a small number produce antibodies that can neutralize either endogenous EPO and recombinant proteins.8484. Casadevall N. Antibodies against rHuEPO: native and recombinant. Nephrol Dial Transplant.2002;17(S5): 42-7.

Most cases of antibody production have been associated with the formulation of epoetin alfa when administered subcutaneously.8585. Macdougall IC, Roger SD, Francisco A, Goldsmith DJA, Schellekens H, Ebbers H, et al. Antibody-mediated pure red cell aplasia in chronic kidney disease patients receiving erythropoiesis-stimulating agents: new insights. Kidney Int.2012;81(8):727-32.

In some cases, the anti-erythropoietin (anti-EPO) anti-body production can lead to development of serious PRCA and transfusion-dependent anemia.86-88 Recent studies have shown that anti-EPO antibody-mediated PRCA is a rare but important adverse effect in patients with CKD who take rHuEPO.89-91

According to the National Guidelines published by Brazilian Ministry of Health, PRCA should be evaluated in patients receiving epoetin alfa over at least four weeks that develop: (1) a drop in hemoglobin levels equal to or greater than 0.5 g/dL per week, in the absence of transfusions, and requirement of at least one red blood cell unit per week to maintain hemoglobin levels; (2) normal leukocyte and platelet counts; (3) absolute reticulocyte count <10 x 103 /µL.9292. Brasil, Ministério da Saúde Protocolo Clínico e Diretrizes Terapêuticas. Aplasia pura adquirida crônica da série vermelha. Brasília: Ministério da Saúde; 2010. Treatment recommendations for patients with PRCA induced by erythropoiesis stimulating agents (ESA) are: (1) discontinuation of ESA; (2) correction of anemia by blood transfusion, if necessary; (3) kidney transplant and (4) introduction of immunosuppressive therapy starting with cyclosporine A alone or in combination with corticosteroids or corticosteroids with cyclophosphamide.8585. Macdougall IC, Roger SD, Francisco A, Goldsmith DJA, Schellekens H, Ebbers H, et al. Antibody-mediated pure red cell aplasia in chronic kidney disease patients receiving erythropoiesis-stimulating agents: new insights. Kidney Int.2012;81(8):727-32. , 8686. Casadevall N. Pure red cell aplasia and anti-erythropoietin antibodies in patients treated with epoetin. Nephrol Dial Transplant. 2003;18 Suppl. 8:37-41.

Diagnostic confirmation of PRCA induced by anti-EPO anti-bodies should include the antibody laboratory detection and a bone marrow examination that shows the absence of erythroid lineage precursors.9393. Casadevall N. What is antibody-mediated pure red cell aplasia (APASV). Nephrol Dial Transplant.2005;20(S4):iv3-8. However, to date, there is no consensus about the method of choice for the detection of these antibodies, since the different methods have advantages and disadvantages.8787. Thorpe R, Swanson SJ. Assays for detecting and diagnosing antibody-mediated pure red cell aplasia (PCRA): an assessment of available procedures. Nephrol Dial Transplant.2005;20(S4):iv16-22. Moreover, no commercial laboratory kit is available for the detection of anti-EPO antibodies in the clinical practice.8989. Shin S, Moon SJ, Ha SK, Jo Y, Lee T, Lee YS, et al. Immunogenicity of recombinant human erythropoietin in Korea: a two-year cross-sectional study. Biologicals. 2012;40(4):254-61.

Genetic polymorphisms and the EPO receptor

Some genetic polymorphisms may result in individual response variations to rHuEPO. Jeong et al.9494. Jeong K, Lee T, Ihm C, Lee S, Moon J. Polymorphisms in two genes, IL-1 B and ACE, are associated with erythropoietin resistance in Korean patients on maintenance hemodialysis. Exp Mol Med. 2008;40(2):161-6. investigated the association between the interleukin 1B (IL-1B) gene and ACE gene polymorphisms and erythropoietin resistance index (RI-EPO) in patients undergoing hemodialysis. Associations between the presence of the IL-1B-511C/C and ACE D/D genotypes with lower RI-EPO were identified, indicating that these polymorphisms may be useful genetic markers in assessing the required dose of rHuEPO in patients undergoing hemodial- ysis.

It is known that endogenous and recombinant EPO stimulates erythropoiesis by binding to the EpoR receptor.9595. Middleton SA, Barbone FP, Johnson DL. Shared and unique determinants of the erythropoietin (EPO) receptor are important for binding EPO and EPO mimetic peptide. J Biol Chem.1999;74(20):14163-9. , 9696. Ng T, Marx G, Littlewood T, Macdougall I. Recombinant erythropoietin in clinical practice. Postgrad Med J. 2003;79(933):367-76. An mRNA alternative splicing can give rise to the soluble form of the receptor (sEpoR) which lacks the transmembrane domain.

sEpoR has a higher affinity for EPO and acts as a potent antagonist of the hormone, which can lead to resistance to treatment with rHuEPO. Therefore, high levels of sEpoR may be associated with administration of high doses of rHuEPO.9797. Inrig J, Bryskin S, Patel U, Arcasoy M, Szczech L. Association between high-dose erythropoiesis stimulating agents, inflammatory biomarkers, and soluble erythropoietin receptors. BMC Nephrol. 2011;12:67. , 9898. Khankin E, Mutter W, Tamez H, Yuan H, Karumanchi S, Thadhani R. Soluble erythropoietin receptor contributes to erythropoietin resistance in end-stage renal disease. PLoS ONE. 2010;5(2):e9246.

Conclusion

The main cause of inadequate response to treatment with rHuEPO is iron deficiency. However, several other factors may be associated with this resistance in patients with CKD on dialysis in the presence of adequate iron stores, and must be investigated including: inflammation, infection, malnutrition, inadequate dialysis and hyperparathyroidism. In addition, B19 infection, anti-EPO antibody production, and the presence of polymorphisms have been identified as possible causes of resistance to rHuEPO in dialysis patients. However, other studies in the Brazilian population, which has its own genetic characteristics, with a larger sample size should be performed to validate these factors.

Finally, as the main reason for poor response to the use of rHuEPO is iron deficiency, which causes changes in the size and color of red blood cells, the importance of monitoring patients under dialysis through a simple blood test should be emphasized, as this may reveal morphological changes such as microcytosis and hypochromia consequent to the deficiency of this nutrient. On the other hand, deficiencies of folic acid and/or vitamin B12 also lead to clinically significant morphological changes in red blood cells, such as macrocytosis. Detection of morphological changes of red blood cells may influence proper supplementation with the adequate nutrient bringing benefits to patients under hemodialysis by properly correcting the anemia.

Acknowledgments

The authors thank FAPEMIG, CNPq/Brazil, and PRPq/UFMG for the financial support. MGC, APF, LMSD and KBG are grateful for CNPq Research grants (PQ).

REFERENCES

  • 1
    Romão Junior JE. A doença renal crônica: do diagnóstico ao tratamento. Prática Hospitalar. 2007;52:183-7.
  • 2
    Manual de Transplante Renal. Associação Brasileira de Transplantes de Órgãos (ABTO). Available from: http://www.abto.org.br/abtov03/Upload/file/ProfissionalManual/manualtransplanterim.pdf [cited 31.10.14].
    » http://www.abto.org.br/abtov03/Upload/file/ProfissionalManual/manualtransplanterim.pdf
  • 3
    Censo da Sociedade Brasileira de Nefrologia 2013. São Paulo: Sociedade Brasileira de Nefrologia (SBN). Available from: http://www.sbn.org.br/pdf/censo2013-14-05.pdf [cited 31.10.14].
    » http://www.sbn.org.br/pdf/censo2013-14-05.pdf
  • 4
    Kazmi WH, Kausz AT, Khan S, Abichandani R, Ruthazer R, Obrador GT, et al. Anemia: an early complication of chronic renal insufficiency. Am J Kidney Dis. 2001;38(4):803-12.
  • 5
    National Kidney Foundation. K/DOQI clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney disease. Am J Kidney Dis.2006;26 Suppl. 3:1-145.
  • 6
    Macdougall IC, Hutton RD, Coles GA, William JD. The use of erythropoietin in renal failure. Postgrad Med J. 1991;67(783):9-15.
  • 7
    Hutchinson FN, Jones WJ. A cost-effectiveness analysis of anemia screening before erythropoietin in patients with endstage renal disease. Am J Kidney Dis.1997;29(5):651-7.
  • 8
    Sesso RC, Lopes AA, Thome FS, Lugon JR, Watanabe Y, Santos DR. Relatório do Censo Brasileiro de Diálise 2012. J Bras Nefrol. 2014;36(1):48-53.
  • 9
    Locatelli F, Aljama P, Barany P, Canaud B, Carrera F, Eckardt KU, et al. Revised European best practice guidelines for the management of anaemia in patients with chronic renal failure. Nephrol Dial Transplant. 2004;19 Suppl. 2:ii1-47.
  • 10
    KDOQI. KDOQI Clinical Practice Guideline and Clinical Practice Recommendations for anemia in chronic kidney disease: 2007 update of hemoglobin target. Am J Kidney Dis.2007;50(3):471-530.
  • 11
    Locatelli F, Covic A, Eckardt KU, Wiecek A, Vanholder R, ERA-EDTA ERBP Advisory Board. Anaemia management in patients with chronic kidney disease: a position statement by the Anaemia Working Group of European Renal Best Practice (ERBP). Nephrol Dial Transplant.2009;24(2):348-54.
  • 12
    KDIGO. KDIGO clinical practice guideline for anemia in chronic kidney disease. Kidney Int Suppl. 2012;2:279-335.
  • 13
    , Locatelli F Pisoni RL, Combe C, Bommer J, Andreucci VE, Piera L, et al. Anaemia in hemodialysis 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.
  • 14
    Brasil. Ministério da Saúde Protocolo Clínico e Diretrizes Terapêuticas. Anemia em pacientes com insuficiência renal crônica - alfaepoetina. Brasília: Ministério da Saúde; 2010.
  • 15
    Ribeiro-Alves MA, Gordan PA. Diagnóstico de Anemia em Pacientes Portadores de Doença Renal Crônica. J Bras Nefrol.2014;36 1 Suppl. 1:9-12.
  • 16
    Abensur H. Anemia da doenc¸a renal crônica. J Bras Nefrol.2004;26(3):26-8.
  • 17
    Brugnara C. Iron deficiency and erythropoiesis: new diagnostic approaches. Clin Chem. 2003;49(10):1573-8.
  • 18
    Mittman N, Sreedhara R, Mushnick R, Chattopadhyay J, Zelmanovic D, Vaseghi M, et al. Reticulocyte hemoglobin content predicts functional iron deficiency in hemodialysis patients receiving rHuEPO. Am J Kidney Dis. 1997;30(6):912-22.
  • 19
    Mast AE, Blinder MA, Dietzen DJ. Reticulocyte hemoglobin content. Am J Hematol. 2008;83(4):307-10.
  • 20
    Fishbane S, Shapiro W, Dutka P, Valenzuela OF, Faubert J. A randomized trial of iron deficiency testing strategies in hemodialysis patients. Kidney Int. 2001;60(6):2406-11.
  • 21
    Brugnara C, Laufer MR, Friedman AJ, Bridges K, Plat O. Reticulocyte hemoglobin content (CHr): early indicator of iron deficiency and response to therapy. Blood. 1994;83(10):3100-1.
  • 22
    Thomas C, Thomas L. Biochemical markers and hematologic indices in the diagnosis of functional iron deficiency. Clin Chem. 2002;48(7):1066-76.
  • 23
    Lea JP, Norris K, Agodoa L. The role of anemia management in improving outcomes for African Americans with chronic kidney disease. Am J Nephrol. 2008;28(5):732-43.
  • 24
    Kalantar-zadeh K, Ikizler TA, Block G, Avram MM, Kopple JD. Malnutrition-inflammation complex syndrome in dialysis patients: causes and consequences. Am J Kidney Dis. 2003;42(5):864-81.
  • 25
    Bamgbola O. Resistance to erythropoietin-stimulating agents: etiology, evaluation, and therapeutic considerations. Pediatr Nephrol. 2010;27(2):195-205.
  • 26
    Wish JB. Assessing iron status: beyond serum ferritin and transferrin saturation. Clin J Am Soc Nephrol. 2006; Suppl. 1:S4-8.
  • 27
    Means RT, Krantz SB. Progress in understanding the pathogenesis of the anemia of chronic disease. Blood.1992;80(7):1639-47.
  • 28
    Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B, Brissot P, et al. A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem. 2001;276(11):7811-9.
  • 29
    Rivera S, Nemeth E, Gabayan V, Lopez MA, Farshidi D, Ganz T. Synthetic hepcidin causes rapid dose-dependent hipoferremia and is concentrated in ferroportin-containing organs. Blood.2005;106(6):2196-9.
  • 30
    Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004;306(5704):2090-3.
  • 31
    De Domenico I, Ward DM, Kaplan J. Hepcidin regulation: ironing out the details. J Clin Invest. 2007;117(7): 1755-8.
  • 32
    , Nemeth E. Ganz T The role of hepcidin in iron metabolism. Acta Haematol. 2009;122(2-3):78-86.
  • 33
    Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK, et al. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest.2004;113(9):1271-6.
  • 34
    , Ganz T. Nemeth E Hepcidin and iron homeostasis. Biochim Biophys Acta. 2012;1823(9):1434-43.
  • 35
    Cançado RD, Chiattone CS. Anemia de Doença Crônica. Rev Bras Hematol Hemoter. 2002;24(2):127-36.
  • 36
    Macdougall IC, Cooper AC. Erythropoietin resistance: the role of inflammation and pro-inflammatory cytokines. Nephrol Dial Transplant. 2002;17(S11):39-43.
  • 37
    Jurado RL. Iron, infections, and anemia of inflammation. Clin Infect Dis. 1997;25(4):888-95.
  • 38
    Fuchs D, Hausen A, Reibnegger G, Werner ER, Werner-felmayer G, Dierich MP, et al. Immune activation and the anaemia associated with chronic inflammatory disorders. Eur J Haematol. 1991;46(2):65-70.
  • 39
    Betjes MG, Huisman M, Weimar W, Litjens NHR. Expansion of cytolytic CD4+ CD28− T cells in end-stage renal disease. Kidney Int.2008;74(6):760-7.
  • 40
    Cooper AC, Breen CP, Vyas B, Ochola J, Kemeny DM,. Macdougall IC Poor response to recombinant erythropoietin is associated with loss of T-lymphocyte CD28 expression and altered interleukin-10 production. Nephrol Dial Transplant.2003;18(1):133-40.
  • 41
    Betjes MG, Weimar W,. Litjens NHR CMV seropositivity determines epoetin dose and hemoglobin levels in patients with CKD. J Am Soc Nephrol. 2009;20(12):2661-6.
  • 42
    Brown KE, Anderson SM, Young NS. Erythrocyte P antigen: cellular receptor for B19 parvovirus. Science.1993;262(5130):114-7.
  • 43
    Chisaka H, Morita E, Yaegashi N, Sugamura K. Parvovirus B19 and the pathogenesis of anaemia. Rev Med Virol. 2003;13(6):347-59.
  • 44
    Hsu C, Bates DW, Kuperman GJ, Curhan GC. Relationship between hematocrit and renal function in men and women. Kidney Int. 2001;59(2):725-31.
  • 45
    Lui SL, Luk WK, Cheung CY, Chan TM, Lai KN, Peiris JS. Nosocomial outbreak of parvovirus b19 infection in a renal transplant unit. Transplantation. 2001;71(1):59-64.
  • 46
    Ozeki M, Fukushima T, Ohzeki M, Sasaki T, Kashihara N. A nosocomial parvovirus B19 infection-induced transient aplastic crisis in a patient with chronic renal failure. Clin Nephrol. 2006;65(2):141-3.
  • 47
    , Young NS. Brown KE Parvovirus B19. N Engl J Med. 2004;350(6):586-97.
  • 48
    Becker MR, Schneider B, Reber U, Poge U, Klein B, Klehr HU, et al. Renal anemia aggravated by long-term parvovirus B19 and cytomegalovirus infection in a renal transplant patient: case report and evaluation of B19 seroprevalence in dialysis patients. Transplant Proc. 2005;37(10):4306-8.
  • 49
    Guiserix J, Ramdane M, Hoarau JM, Finielz P, Michault A. Parvovirus B19 and hemodialysis. Nephron. 1996;72(4): 719.
  • 50
    Malyszko J, Hryszko T, Malyszko JS, Wolczynski S, Mysliwiec M. Parvovirus B19 infection and IGF system components in relation to erythropoiesis in dialyzed patients and kidney transplant recipients. Transplant Proc.2002;34(8):3211-4.
  • 51
    Duranay M, Bali M, Sahin M, Yakinci G, Vurgun N, Dilmen U. Parvovirus B19 infection and unresponsiveness to erythropoietin therapy in haemodialysis patients. Nephrol Dial Transplant.1998;13(3):779-80.
  • 52
    Alves MT, Vilac¸a SS, Carvalho MG, Fernandes AP, Dusse LM, Gomes KB. Human parvovirus B19 infection in a renal transplant recipient: a case report. BMC Res Notes. 2013; 6(28).
  • 53
    Egbuna O, Zand MS, Arbini A, Menegus M, Taylor J. A cluster of parvovirus B19 infections in renal transplant recipients: a prospective case series and review of the literature. Am J Transplant. 2006;6(1):225-31.
  • 54
    Smith SR, Butterly DW, Alexander BD, Greenberg A. Viral infections after renal transplantation. Am J Kidney Dis.2001;137(4):659-76.
  • 55
    Keung Y, Chuahirun T, Wesson D. Concomitant parvovirus b19 and cytomegalovirus infections after living-related renal transplantation. Nephrol Dial Transplant.1999;14(2):469-71.
  • 56
    Fish P, Handgretinger R, Shaefer H. Pure red cell aplasia. Br J Haematol. 2000;111(4):1010-22.
  • 57
    Avesani CM, Carrero JJ, Axelsson J, Qureshi AR, Lindholm B, Stenvinkel P. Inflammation and wasting in chronic kidney disease: partners in crime. Kidney Int.2006;70:8-13.
  • 58
    Qureshi AR, Alvestrand A, Danielsson A, Divino-Filho JC, Gutierrez A, Lindholm B, et al. Factors predicting malnutrition in hemodialysis patients: a cross-sectional study. Kidney Int.1998;53(3):773-82.
  • 59
    , Locatelli F Andrulli S, Memoli B, Maffei C, Vecchio LD, Aterini S, et al. Nutritional-inflammation status and resistance to erythropoietin therapy in haemodialysis patients. Nephrol Dial Transplant.2006;21:991-8.
  • 60
    Kalantar-Zadeh K, Lee GH, Miller JE, Streja E, Jing J, Robertson JA, et al. Predictors of hyporesponsiveness to erythropoiesis-stimulating agents in hemodialysis patients. Am J Kidney Dis.2009;53(5):823-34.
  • 61
    Gaweda AE, Goldsmith LJ, Brier ME, Aronoff GR. Iron, inflammation, dialysis adequacy, nutritional status, and hyperparathyroidism modify erythropoietic response. Clin J Am Soc Nephrol. 2010;5(4):576-81.
  • 62
    Shaefer R, Teschner M, Kosch M. Folate metabolism in renal failure. Nephrol Dial Transplant.2002;17(S5):24-7.
  • 63
    Perna AF, Ingrosso D, Santo NG, Galletti P, Brunone M, Zappia V. Metabolic consequences of folate-induced reduction of hyperhomocysteinemia in uremia. Am Soc Nephrol. 1997;8(12):1899-905.
  • 64
    Vecchi AF, Bamonti-Catena F, Finazzi S, Campolo J, Taioli E, Novembrino C, et al. Homocysteine, vitamin b12, and serum and erythrocyte folate in peritoneal dialysis and hemodialysis patients. Perit Dial Int. 2000;20(2):169-73.
  • 65
    Costa E, Belo L, Santos-Silva A. Special problems in hemodialysis patients Internet. In: Resistance to recombinant human erythropoietin therapy in haemodialysis patients. Portugal: In Tech; 2011 [Chapter 3]. Available from: http://www.intechopen.com/books/special-problems-in-hemodialysis-patients/resistance-to-recombinant-human-erythropoietin-therapy-in-haemodialysis-patients [cited 14.10.13].
    » http://www.intechopen.com/books/special-problems-in-hemodialysis-patients/resistance-to-recombinant-human-erythropoietin-therapy-in-haemodialysis-patients
  • 66
    Shahab I, Khanna R, Nolph KD. Peritoneal dialysis or hemodialysis? A dilemma for the nephrologist. Adv Perit Dial. 2006;22:180-5.
  • 67
    National Kidney Foundation. K/DOQI clinical practice guidelines for nutrition in chronic renal failure. Am J Kidney Dis.2000;35 6 Suppl. 2:S1-40.
  • 68
    Movilli E, Cancarini GC, Zani R, Camerini C, Sandrini M, Maiorca R. Adequacy of dialysis reduces the doses of recombinant erythropoietin independently from the use of biocompatible membranes in haemodialysis patients. Nephrol Dial Transplant.2001;16(1):111-4.
  • 69
    Druek TB, Eckard K. Role of secondary hyperparathyroidism in erythropoietin resistance of chronic renal failure patients. Nephrol Dial Transplant.2002;17(S5):28-31.
  • 70
    Brancaccio D, Cozzolino M, Gallieni M. Hyperparathyroidism and anemia in uremic subjects: a combined therapeutic approach. J Am Soc Nephrol. 2004;15 Suppl. 1:S21-4.
  • 71
    National Kidney Foundation. Kidney disease-dialysis outcome quality initiative: K/DOQI Clinical Practice Guidelines for bone metabolism and disease in chronic kidney disease. Am J Kidney Dis.2003;42 4 Suppl. 3:S1-202.
  • 72
    Rao DS, Shih M, Mohini R. Effect of serum parathyroid hormone and bone marrow fibrosis on the response to erythropoietin in uremia. N Engl J Med.1993;328(3):171-5.
  • 73
    Vlahakos DV, Marathias KP, Madias NE. The role of the renin-angiotensin system in the regulation of erythropoiesis. Am J Kidney Dis.2010;56(3):558-65.
  • 74
    Rodgers KE, diZerega JS. Contribution of the local RAS to hematopoietic function: a novel therapeutic target. Front Endocrinol (Lausanne). 2013;23(4):157.
  • 75
    Erdös EG. Angiotensin I converting enzyme and the changes in our concepts through the years. Hypertension. 1990;16(4):363-70.
  • 76
    Lenfant M, Wdzieczak-bakala J, Guittet E, Prome JC, Sotty D, Frindel E. Inhibitor of hematopoietic pluripotent stem cell proliferation: purification and determination of its structure. Proc Natl Acad Sci USA. 1989;86(3):779-82.
  • 77
    Rieger K, Saez-servent N, Papet MP, Wdzieczak-Bakala J, Morgat J, Thierry J, et al. Involvement of human plasma angiotensin I-converting enzyme in the degradation of the haemoregulatory peptide N-acetyl seryl aspartyl lysyl proline. Biorem J. 1993;296(Pt. 2):373-8.
  • 78
    Azizi M, Rousseau A, Ezan E, Guyene T, Michelet S, Grognet J, et al. Acute angiotensin-converting enzyme inhibition increases the plasma level of the natural stem cell regulator N-acetyl-seryl-aspartyl-lysyl-proline. J Clin Invest. 1991;97(3):839-44.
  • 79
    Meur YL, Lorgeot V, Comte L, Szelaq JC, Leroux-Robert C, Praloran V. Plasma levels and metabolism of AcSDKP in patients with chronic renal failure: relationship with erythropoietin requirements. Am J Kidney Dis.2001;38(3):510-7.
  • 80
    Cole J, Ertoy D, Lin H, Sutliff RL, Ezan E, Guyene TT, et al. Lack of angiotensin II - facilitated erythropoiesis causes anemia in angiotensin-converting enzyme-deficient mice. J Clin Invest.2000;106(11):1391-8.
  • 81
    Iodice C, Balletta MM, Minutolo R, Giannattasio P, Tuccillo S, Bellizzi V, et al. Maximal suppression of renin-angiotensin system in nonproliferative glomerulonephritis. Kidney Int.2003;63(6):2214-21.
  • 82
    Jacobsen P, Andersen S, Jensen BR, Parving HH. Additive effect of ACE inhibition and angiotensin II receptor blockade in type I diabetic patients with diabetic nephropathy. J Am Soc Nephrol. 2003;14(4):992-9.
  • 83
    Marathias KP, Agroyannis B, Mavromoustakos T, Matsoukas J,. Vlahakos DV Hematocrit-lowering effect following inactivation of renin-angiotensin system with angiotensin converting enzyme inhibitors and angiotensin receptor blockers. Curr Top Med Chem. 2004;4(4):483-6.
  • 84
    Casadevall N. Antibodies against rHuEPO: native and recombinant. Nephrol Dial Transplant.2002;17(S5): 42-7.
  • 85
    Macdougall IC, Roger SD, Francisco A, Goldsmith DJA, Schellekens H, Ebbers H, et al. Antibody-mediated pure red cell aplasia in chronic kidney disease patients receiving erythropoiesis-stimulating agents: new insights. Kidney Int.2012;81(8):727-32.
  • 86
    Casadevall N. Pure red cell aplasia and anti-erythropoietin antibodies in patients treated with epoetin. Nephrol Dial Transplant. 2003;18 Suppl. 8:37-41.
  • 87
    Thorpe R, Swanson SJ. Assays for detecting and diagnosing antibody-mediated pure red cell aplasia (PCRA): an assessment of available procedures. Nephrol Dial Transplant.2005;20(S4):iv16-22.
  • 88
    Behler C, Terrault N, Etzell J, Damon L. Rituximab therapy for pure red cell aplasia due to anti-epoetin antibodies in a woman treated with epoetin-alfa: a case report. J Med Case Rep. 2009;3:7335.
  • 89
    Shin S, Moon SJ, Ha SK, Jo Y, Lee T, Lee YS, et al. Immunogenicity of recombinant human erythropoietin in Korea: a two-year cross-sectional study. Biologicals. 2012;40(4):254-61.
  • 90
    Kharagjitsingh AV, Korevaar JC, Vandenbroucke JP, Boeschoten EW, Krediet RT, Daha MR, et al. Incidence of recombinant erythropoietin (EPO) hyporesponse, EPO-associated antibodies, and pure red cell aplasia in dialysis patients. Kidney Int.2005;68(3):1215-22.
  • 91
    Schonholzer C, Keusch G, Nigg L, Robert D, Wauters J. High prevalence in Switzerland of pure red-cell aplasia due to anti-erythropoietin antibodies in chronic dialysis patients: report of five cases. Nephrol Dial Transplant.2004;19(8):2121-5.
  • 92
    Brasil, Ministério da Saúde Protocolo Clínico e Diretrizes Terapêuticas. Aplasia pura adquirida crônica da série vermelha. Brasília: Ministério da Saúde; 2010.
  • 93
    Casadevall N. What is antibody-mediated pure red cell aplasia (APASV). Nephrol Dial Transplant.2005;20(S4):iv3-8.
  • 94
    Jeong K, Lee T, Ihm C, Lee S, Moon J. Polymorphisms in two genes, IL-1 B and ACE, are associated with erythropoietin resistance in Korean patients on maintenance hemodialysis. Exp Mol Med. 2008;40(2):161-6.
  • 95
    Middleton SA, Barbone FP, Johnson DL. Shared and unique determinants of the erythropoietin (EPO) receptor are important for binding EPO and EPO mimetic peptide. J Biol Chem.1999;74(20):14163-9.
  • 96
    Ng T, Marx G, Littlewood T, Macdougall I. Recombinant erythropoietin in clinical practice. Postgrad Med J. 2003;79(933):367-76.
  • 97
    Inrig J, Bryskin S, Patel U, Arcasoy M, Szczech L. Association between high-dose erythropoiesis stimulating agents, inflammatory biomarkers, and soluble erythropoietin receptors. BMC Nephrol. 2011;12:67.
  • 98
    Khankin E, Mutter W, Tamez H, Yuan H, Karumanchi S, Thadhani R. Soluble erythropoietin receptor contributes to erythropoietin resistance in end-stage renal disease. PLoS ONE. 2010;5(2):e9246.

Publication Dates

  • Publication in this collection
    May-Jun 2015

History

  • Received
    23 July 2014
  • Accepted
    24 Nov 2014
Associação Brasileira de Hematologia e Hemoterapia e Terapia Celular R. Dr. Diogo de Faria, 775 cj 114, 04037-002 São Paulo/SP/Brasil, Tel. (55 11) 2369-7767/2338-6764 - São Paulo - SP - Brazil
E-mail: secretaria@rbhh.org