Your activity: 28 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Pure red cell aplasia (PRCA) due to anti-erythropoiesis-stimulating agent antibodies

Pure red cell aplasia (PRCA) due to anti-erythropoiesis-stimulating agent antibodies
Author:
Jeffrey S Berns, MD
Section Editor:
Steve J Schwab, MD, FACP, FASN
Deputy Editor:
Eric N Taylor, MD, MSc, FASN
Literature review current through: Dec 2022. | This topic last updated: Sep 14, 2021.

INTRODUCTION — Acquired pure red cell aplasia (PRCA) is a rare condition of profound anemia characterized by a very low reticulocyte count and the virtual absence of erythroid precursors in the bone marrow. All other cell lines are present and seem quantitatively and morphologically normal. Many cases of acquired PRCA are idiopathic. In others, underlying conditions, such as thymoma, myelodysplastic syndromes, lymphoma, leukemia, systemic autoimmune disorders, and viral infection (ie, parvovirus B19), or treatment with drugs, such as phenytoin or chloramphenicol, are identified [1]. (See "Acquired pure red cell aplasia in adults".)

PRCA has also been described in patients treated with epoetin and other erythropoiesis-stimulating agents (ESAs), resulting from the induction of neutralizing antibodies directed against the erythropoietin (EPO) molecule [2]. Most reported cases have been in patients receiving epoetin for chronic kidney disease (CKD)-related anemia.

PRCA due to anti-EPO antibodies in patients with CKD will be reviewed here. A discussion of other causes of acquired PRCA is presented separately. (See "Acquired pure red cell aplasia in adults".)

ETIOLOGY AND PATHOGENESIS — Most cases of non-erythropoietin (EPO)-related PRCA are mediated by immunoglobulin G (IgG) autoantibodies or cytotoxic T lymphocytes directed against erythroid precursor or progenitor cells [1,3]. Inhibition of erythropoiesis occurs primarily between the colony forming unit–erythroid (CFU-E) and proerythroblast stages. The proerythroblast is the first morphologically identifiable erythroid precursor in normal marrow and is virtually absent in the marrow of patients with PRCA. (See "Acquired pure red cell aplasia in adults".)

PRCA due to autoantibodies against endogenous EPO is rare in patients who have never been treated with erythropoiesis-stimulating agents (ESAs) [4-6]. The serum of patients with EPO-related PRCA inhibits the growth of erythroid progenitor cells in bone marrow cultures [2]. Neutralizing IgG antibodies to the protein component of exogenous ESAs crossreact with endogenous EPO.

There are several preparations of epoetins (ie, epoetin alfa, epoetin beta, epoetin omega, and others) produced by various manufacturers around the world. They differ from one another and from the native hormone in terms of glycosylation and sialic acid content [7]. The vast majority of cases of EPO-related PRCA have occurred in patients treated with a particular epoetin alfa product (Eprex, in single-use syringes), which was manufactured and distributed outside the United States. Altered antigenicity of this specific product has been suggested as the underlying cause of anti-EPO antibody development. However, EPO-related PRCA has been described with other preparations including darbepoetin alfa and methoxy polyethylene glycol-epoetin beta [8,9].

Virtually all reported cases of anti-EPO antibody-mediated PRCA have occurred in patients with chronic kidney disease (CKD) who have received the drug subcutaneously [2,10-12]. Although not specifically studied with epoetin, subcutaneous administration of other drugs has been associated with greater immunogenicity than with intravenous (IV) administration [13].

EPIDEMIOLOGY — Most cases of erythropoietin (EPO)-related PRCA occurred in patients with renal failure in Europe and the United Kingdom, but patients in Canada, Australia, and countries in Asia have also been affected [10,14].

The condition remains extremely rare, given the widespread use of epoetin and other erythropoiesis-stimulating agents (ESAs) [15-17]. The overall incidence of reported cases between 1989 and June 2004 was 1.6 per 10,000 patient-years of subcutaneous exposure [18]. There have been only a few cases reported in patients treated with intravenously (IV) administered epoetin, with an incidence of 0.02 per 10,000 patient-years of exposure [19].

Eprex — There have been over 200 reported cases of PRCA related to Eprex use [20], representing the vast majority of affected patients [10,14]. There was a substantial increase in the number of reported cases between 2001 and 2003, almost exclusively in patients with chronic kidney disease (CKD) who received Eprex subcutaneously [10,14,18].

This increase in the number of cases was later discovered to coincide with the release of a new formulation and packaging of this epoetin, as discussed above. The incidence was 3.43 per 10,000 patient-years of exposure for Eprex supplied in syringes with uncoated rubber stoppers compared with 0.23 and 0.17 per 10,000 patient-years for Eprex preparations in coated rubber-stopper syringes and stabilized with human serum albumin (HSA), respectively.

One report of the multinational Prospective Immunogenicity Surveillance Registry (PRIMS) described five confirmed episodes of EPO-related PRCA [21]. Three episodes occurred with a reformulated Eprex preparation, and two occurred with other ESAs among over 15,000 patients, with an incidence (per 100,000 patient-years) of 35.8 for Eprex and 14 for other ESAs (rate ratio 2.56, 95% CI 0.43-15.31), which was not statistically significantly different.

Non-Eprex cases — The overall incidence of reported cases between 1989 and 2004 of PRCA with subcutaneous use of non-Eprex preparations was much lower, ranging from 0.02 to 0.16 per 10,000 patient-years between 1989 and 2004, depending on the preparation [10,14,17,22-26]. Fortunately, EPO PRCA has become exceedingly rare since 2005 [26].

EPO antibody-mediated PRCA has only rarely been reported in children with kidney failure [14,27].

"Biosimilar" epoetin products are becoming increasingly available around the world; treatment with these agents has also been associated with development of anti-EPO both non-neutralizing and neutralizing antibodies, with a notable outbreak in Thailand related to subcutaneous use of a biosimilar produced outside the United States and the European Union [28-31].

SCREENING FOR ANTI-EPO ANTIBODIES — Among dialysis patients, routine screening for the presence of anti-erythropoietin (EPO) antibodies cannot be justified. This was shown in a study involving five centers in Canada, in which approximately 1500 hemodialysis, peritoneal dialysis, and predialysis patients were screened [32]. Using a radioimmunoprecipitation assay (RIPA), a low-positive test was noted in only one patient (who had previously been diagnosed with PRCA), and borderline positive assays were observed in only three individuals, none of whom had signs of PRCA. None of the four had neutralizing anti-EPO antibodies.

Similar results were obtained in 536 patients screened in 35 German dialysis units [33]. In this study, anti-EPO antibodies were detected in three EPO-hyporesponsive and three EPO-normoresponsive patients using an enzyme-linked immunosorbent assay (ELISA), with only one borderline result with a RIPA. There were no cases of PRCA.

However, we agree with the 2012 Kidney Disease: Improving Global Outcomes (KDIGO) guidelines, which suggest that an evaluation for PRCA due to anti-EPO antibodies should occur in the patient exposed to at least eight weeks of erythropoiesis-stimulating agent (ESA) therapy who develops all of the following [34]:

Decline in hemoglobin (Hb) level of >0.5 to 1 g/dL per week or transfusion requirement of at least one to two units per week to maintain adequate Hb

Normal platelet and white blood cell (WBC) count

Absolute reticulocyte count of <10,000/microL

CLINICAL MANIFESTATIONS — PRCA due to anti-erythropoietin (EPO) antibodies should be suspected if the hemoglobin (Hb) level declines by >2 g/dL per month or the reticulocyte count is <20,000/microL in an individual who has previously responded to EPO. PRCA generally does not occur, unless the patient has been on EPO for at least three to four weeks, and typically occurs after 6 to 18 months of exposure [10].

PRCA is specifically characterized by the following clinical features [10,35]:

Decline in Hb level of >0.7 to 1 g/dL per week without transfusions or transfusion requirement of at least one unit per week to maintain adequate Hb, despite continued use of EPO at the same or increased doses

Markedly reduced reticulocyte count (<10,000/microL)

Drop in platelet counts, although generally not below the lower limit of normal

Normal white blood cell (WBC) count

Elevated serum transferrin saturation and serum ferritin, reflecting decreased utilization of iron secondary to diminished erythropoiesis

Allergic urticarial skin reactions at sites of earlier subcutaneous EPO injections have also been described [36].

EVALUATION AND DIAGNOSIS — To properly diagnose the disorder, a bone marrow aspirate and evaluation for the presence of anti-erythropoietin (EPO) antibodies must be performed.

Bone marrow aspirate — The bone marrow aspirate reveals severe erythroid hypoplasia, with <5 percent red blood cell precursors; evidence of a block in the maturation of erythroid precursors may be present. Platelet and white cell precursors are entirely normal.

Anti-EPO antibodies — Identification of antibodies to EPO is a critical component of the diagnosis. There are several available tests to detect antibodies that bind EPO, each with its own advantages and disadvantages [37]:

Radioimmunoprecipitation assay (RIPA) appears to be the most accurate test for detecting anti-EPO antibodies. However, it is not standardized, may not detect low-affinity antibodies, requires radiolabeled antigen, and is time consuming and difficult to automate [10].

Enzyme-linked immunosorbent assays (ELISAs) are more widely available but appear to have lower sensitivity and specificity than the RIPA [10,38].

A biosensor assay, which is not as readily available as the previous two assays, may provide better characterization of detected antibodies (ie, binding affinity and isotype). However, it may detect very low-affinity antibodies in baseline sera that never cause PRCA.

In general, these assays can detect antibodies with a concentration of at least 500 ng/mL [37]. The neutralizing capability of the anti-EPO antibodies cannot be assessed with any of the above-mentioned assays. This requires a bioassay in which patient serum or immunoglobulin inhibits red blood cell precursor growth in bone marrow or cell line cultures [10,35]. In addition, non-neutralizing antibodies may be more common than neutralizing antibodies [31].

It is difficult to recommend use of any one of these assays over another given that they are not standardized, have advantages and disadvantages that are difficult to compare, and are not available commercially [39]. The World Health Organization (WHO) established an EPO-antibody reference panel that should be useful for assay standardization and improving diagnostic accuracy [40].

The sample in a suspected case may be sent to the manufacturer, where several different assays, including one for neutralizing capability, are performed [41]. Confirmation of a suspected case of anti-EPO antibody-mediated PRCA should show the presence of anti-EPO antibodies and evidence of neutralizing ability [14,35]. Bioassays generally detect antibodies with a concentration of at least 1 mcg/mL [37].

DIFFERENTIAL DIAGNOSIS — In addition to PRCA due to anti-erythropoietin (EPO) antibodies, there are a large number of other causes of anemia and hyporesponsiveness to EPO among patients with chronic kidney disease (CKD). They can be distinguished from PRCA largely because of the observation that PRCA due to anti-EPO antibodies is associated with a profound fall in hemoglobin (Hb) level and reticulocyte count [37]. By comparison, the other causes of EPO resistance are associated with less severe drops in counts. Other causes of EPO resistance are discussed separately. (See "Hyporesponse to erythropoiesis-stimulating agents (ESAs) in chronic kidney disease", section on 'Causes'.)

TREATMENT — There is limited experience with managing PRCA. Treatment consists of:

Initial management including transfusions for symptomatic anemia and discontinuing all recombinant erythropoietin (EPO) products

Immunosuppressive therapy to eradicate antibodies

Among patients who do not respond to immunosuppressive therapies and are eligible candidates, consideration should be given to kidney transplantation.

Initial management — The two most important initial steps in management in anti-EPO antibody-mediated PRCA are transfusions for symptomatic anemia and stopping all recombinant EPO products [10]. There are at least three observations underlying the recommendation to avoid any recombinant EPO preparation, including darbepoetin alfa, in patients with anti-EPO antibody-mediated PRCA:

Anti-EPO antibodies crossreact not only with the endogenous hormone, but also with all recombinant EPO molecules, including darbepoetin alfa [2].

Rechallenge with EPO preparations may cause an anamnestic antibody response, making it less possible for the antibody to either spontaneously disappear or return to clinically unimportant levels.

Rechallenge with EPO may incite the formation of allergic skin and systemic reactions (eg, anaphylaxis), which might further complicate the patient's clinical picture [36].

However, there have been case reports of patients who have tolerated rechallenge [8,42-47]. Despite these case reports and other isolated instances in which patients appeared to tolerate resumption of EPO therapy or other EPO preparations, in some instances with concomitant immunosuppression [42,48], patients with EPO-associated PRCA should have EPO treatment discontinued and should not routinely be switched to an alternative EPO product or to darbepoetin alfa.

If, however, rechallenge is considered for clinical reasons or at the patient's request, the following practice appears reasonable [45]:

Rechallenge only if anti-EPO antibody levels are below the lower limit of detection

Monitor the absolute reticulocyte count and anti-EPO antibody levels and for systemic reactions

Administer methoxy polyethylene glycol-epoetin beta, rather than EPO or darbepoetin

Administer methoxy polyethylene glycol-epoetin beta intravenously, rather than subcutaneously, whenever feasible

Immunosuppressive therapy — Since PRCA in this setting is immune mediated, the condition should respond to immunosuppressive therapy, and, while spontaneous remissions after cessation of EPO therapy occur, they appear to be rare; immunosuppressive therapy should probably be provided in most cases [45].

There are no controlled data, but several different immunosuppressive regimens have been used. In one retrospective review of 47 cases in Europe [49], EPO therapy was stopped in all patients. Ten patients did not receive immunosuppressive therapy, one died within six weeks of diagnosis, and nine did not recover by a median follow-up of 12 months.

The remaining 37 patients were treated with a variety of immunosuppressive agents. The majority received steroids, alone or in combination with cyclophosphamide, intravenous immune globulin (IVIG), or plasmapheresis; six received cyclosporine alone; one received mycophenolate mofetil; and two received rituximab. Six patients received a kidney transplant. The following results were reported:

Seventy-eight percent of all patients recovered, most within three months.

Recovery occurred only after antibody levels became undetectable.

No relapse of PRCA occurred following cessation of immunosuppression, but none of the patients were rechallenged with EPO or darbepoetin alfa following recovery.

All six patients who received a kidney transplant recovered.

Favorable responses were seen with many of the regimens. This included corticosteroids with or without IVIG (10 of 18 recovered) and corticosteroids with cyclophosphamide (seven of eight recovered), as well as with cyclosporine alone (four of six recovered), which resulted in the fastest response [49,50]. In this study, none of the patients treated with rituximab, an antibody against B cells that has been effective for non-EPO-related PRCA, recovered [49], although response to rituximab has been reported elsewhere [51,52]. Only one of nine patients who were treated with IVIG alone recovered.

Tacrolimus has also been used successfully in a case report [53].

In one review, there was an analysis of EPO-related PRCA, as reported to the US Food and Drug Administration (FDA) and the manufacturers of several EPO preparations, as well as longer-term follow-up of patients previously reported and described above from the European PRCA Study Group [49]. It was concluded that the highest recovery rates were associated with combination cyclophosphamide and prednisone and that the highest rates of EPO responsiveness on rechallenge were in those patients whose anti-EPO antibody levels were undetectable [54].

The treatment recommendations that follow are made with the caveat that worldwide experience is very limited, and none of the data are from randomized, controlled trials. Given that the highest recovery rates appear to be associated with combination cyclophosphamide and prednisone, we prefer initial therapy consisting of prednisone (1 mg/kg per day) plus oral cyclophosphamide (50 to 100 mg per day). A reasonable alternative is cyclosporine alone at a dose of 200 mg per day (or 100 mg twice per day) [45,54]. Patients who do not respond to treatment with either oral cyclophosphamide plus prednisone or with cyclosporine alone may be treated with the other regimen; tacrolimus may also be considered as an alternative to cyclosporine. (See "General principles of the use of cyclophosphamide in rheumatic diseases".)

The cost, potential toxicities, and limited effectiveness [49] of IVIG and rituximab suggest that these treatments should perhaps be limited to those who fail to respond to other therapy. (See "Overview of intravenous immune globulin (IVIG) therapy".)

The optimal duration of immunosuppressive therapy and monitoring for anti-EPO antibody-mediated PRCA is unknown. However, the following recommendations appear reasonable:

Continue treatment until antibody levels become undetectable; discontinue treatment three or four months after initiation among patients with no response.

Monitor hemoglobin (Hb) weekly to assess for transfusion requirement and response to therapy.

Monitor reticulocyte count and anti-EPO antibody levels every one to two weeks during treatment to assess response or lack thereof.

Since the majority of patients who respond to treatment do so within three to four months, immunosuppressive therapy should probably be discontinued if there is no response within this time. However, in the abovementioned study of 37 treated patients, 4 patients responded 5 to 18 months after treatment was initiated [49].

With the introduction of "biosimilar" epoetins and increasing use of unlicensed epoetin products in certain parts of the world, ongoing vigilance must be maintained for new occurrences of anti-EPO antibody-mediated PRCA [31,55].

Experimental therapies — Hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF PHIs) are a novel class of oral drugs that stimulate the production of endogenous erythropoietin. The HIF-PHI roxadustat may be helpful for the management of anemia in patients with EPO-associated PRCA [56-60]. However, these data are limited to case reports and additional data are needed before these can be routinely recommended for management of EPO-related PRCA.

SUMMARY AND RECOMMENDATIONS

The vast majority of cases of recombinant human erythropoietin (EPO)-related acquired pure red cell aplasia (PRCA) have occurred in patients treated with a particular epoetin alfa product: Eprex (in single-use syringes). The underlying cause may be organic compounds (leached by polysorbate from uncoated rubber stoppers in prefilled syringes) that are acting as adjuvants, resulting in anti-EPO antibody development. Virtually all reported cases of anti-EPO antibody-mediated PRCA have occurred in patients with chronic kidney disease who have received the drug subcutaneously. (See 'Etiology and pathogenesis' above.)

EPO-related PRCA remains extremely rare, given the widespread use of erythropoiesis-stimulating agent (ESAs). There have been >200 reported cases of PRCA related to Eprex use, which represent the vast majority of affected patients. Following changes in storage and handling recommendations and discontinuation of subcutaneous administration of Eprex, the incidence of anti-EPO antibody-mediated PRCA in patients with kidney disease has declined. (See 'Epidemiology' above.)

Screening for the presence of anti-EPO antibodies should not be routinely performed in dialysis patients. (See 'Screening for anti-EPO antibodies' above.)

EPO-induced PRCA should be considered in the patient with significant anemia who has been treated with epoetin or another ESA for at least three to four weeks and has previously responded to treatment. The condition is characterized by a sudden decline in hemoglobin (Hb) level despite continued use of the ESA, markedly reduced reticulocyte count, and normal white blood cell (WBC) and platelet counts. (See 'Clinical manifestations' above.)

To definitively diagnose EPO-induced PRCA, a bone marrow aspirate and evaluation for the presence of neutralizing anti-EPO antibodies should be performed. The bone marrow reveals severe erythroid hypoplasia, with <5 percent red blood cell precursors, and there may be evidence of a block in the maturation of erythroid precursors. Platelet and white cell precursors are entirely normal. Anti-EPO antibodies are detected by radioimmunoprecipitation assay (RIPA), enzyme-linked immunosorbent assays (ELISAs), or other assay, as available. The sample must be sent to the manufacturer for testing to document the presence of anti-EPO antibodies and evidence of their neutralizing ability. (See 'Evaluation and diagnosis' above.)

We recommend the cessation of all recombinant ESAs in patients with PRCA (Grade 1A). We also recommend not switching to an alternative ESA (Grade 1A). Patients should be transfused for severe symptomatic anemia. Our recommendations concerning rechallenge are discussed below. (See 'Immunosuppressive therapy' above.)

Given that spontaneous remissions after cessation of epoetin therapy are rare, we recommend the administration of immunosuppressive therapy in most patients (Grade 1B). We suggest initial therapy consisting of prednisone (1 mg/kg per day) plus oral cyclophosphamide (50 to 100 mg per day) for a maximum of three to four months (Grade 2C). A reasonable alternative for first-line therapy is cyclosporine alone at a dose of 200 mg per day (or 100 mg twice per day) for a maximum of three to four months. Patients who do not respond to initial treatment with either oral cyclophosphamide plus prednisone or with cyclosporine alone may be subsequently treated with the other regimen. Tacrolimus may also be considered as an alternative to cyclosporine. Consideration should be given to kidney transplantation in all eligible patients if there is no response to this strategy. (See 'Immunosuppressive therapy' above and "General principles of the use of cyclophosphamide in rheumatic diseases".)

The optimal duration of immunosuppressive therapy and monitoring for anti-EPO antibody-mediated PRCA is unknown. However, the following recommendations appear reasonable (see 'Immunosuppressive therapy' above and "General principles of the use of cyclophosphamide in rheumatic diseases"):

Continue treatment until antibody levels become undetectable; discontinue treatment three or four months after initiation among patients with no response.

Monitor Hb weekly to assess for transfusion requirement and response to therapy.

Monitor absolute reticulocyte count and anti-EPO antibody levels every one to two weeks during treatment to assess response or lack thereof.

Rechallenge may be considered in patients in whom anti-EPO antibody levels are below or closer to the lower limit of detection. If performed, we recommend administration of the methoxy polyethylene glycol-epoetin beta intravenously in hemodialysis patients. The Hb, reticulocyte count, and anti-EPO antibody levels should be closely monitored. (See 'Initial management' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Stanley L Schrier, MD (deceased), who contributed to an earlier version of this topic review.

  1. Fisch P, Handgretinger R, Schaefer HE. Pure red cell aplasia. Br J Haematol 2000; 111:1010.
  2. Casadevall N, Nataf J, Viron B, et al. Pure red-cell aplasia and antierythropoietin antibodies in patients treated with recombinant erythropoietin. N Engl J Med 2002; 346:469.
  3. Krantz S. Anemia due to bone marrow failure: diagnosis and treatment. Compr Ther 1980; 6:10.
  4. Peschle C, Marmont AM, Marone G, et al. Pure red cell aplasia: studies on an IgG serum inhibitor neutralizing erythropoietin. Br J Haematol 1975; 30:411.
  5. Bunn HF. Drug-induced autoimmune red-cell aplasia. N Engl J Med 2002; 346:522.
  6. Casadevall N, Dupuy E, Molho-Sabatier P, et al. Autoantibodies against erythropoietin in a patient with pure red-cell aplasia. N Engl J Med 1996; 334:630.
  7. Storring PL, Tiplady RJ, Gaines Das RE, et al. Epoetin alfa and beta differ in their erythropoietin isoform compositions and biological properties. Br J Haematol 1998; 100:79.
  8. Shingu Y, Nakata T, Sawai S, et al. Antibody-mediated pure red cell aplasia related with epoetin-beta pegol (C.E.R.A.) as an erythropoietic agent: case report of a dialysis patient. BMC Nephrol 2020; 21:275.
  9. Padhi S, Behera G, Pattnaik SA, et al. Acquired Pure Red Cell Aplasia Following Recombinant Erythropoietin (Darbepoetin-alfa) Therapy. Indian J Nephrol 2020; 30:113.
  10. Rossert J, Casadevall N, Eckardt KU. Anti-erythropoietin antibodies and pure red cell aplasia. J Am Soc Nephrol 2004; 15:398.
  11. Eckardt KU, Casadevall N. Pure red-cell aplasia due to anti-erythropoietin antibodies. Nephrol Dial Transplant 2003; 18:865.
  12. Quint L, Casadevall N, Giraudier S. Pure red cell aplasia in patients with refractory anaemia treated with two different recombinant erythropoietins. Br J Haematol 2004; 124:842.
  13. Schellekens H. Immunogenicity of therapeutic proteins: clinical implications and future prospects. Clin Ther 2002; 24:1720.
  14. Bennett CL, Luminari S, Nissenson AR, et al. Pure red-cell aplasia and epoetin therapy. N Engl J Med 2004; 351:1403.
  15. Prabhakar SS, Muhlfelder T. Antibodies to recombinant human erythropoietin causing pure red cell aplasia. Clin Nephrol 1997; 47:331.
  16. Peces R, de la Torre M, Alcázar R, Urra JM. Antibodies against recombinant human erythropoietin in a patient with erythropoietin-resistant anemia. N Engl J Med 1996; 335:523.
  17. Gershon SK, Luksenburg H, Coté TR, Braun MM. Pure red-cell aplasia and recombinant erythropoietin. N Engl J Med 2002; 346:1584.
  18. Boven K, Stryker S, Knight J, et al. The increased incidence of pure red cell aplasia with an Eprex formulation in uncoated rubber stopper syringes. Kidney Int 2005; 67:2346.
  19. Cournoyer D, Toffelmire EB, Wells GA, et al. Anti-erythropoietin antibody-mediated pure red cell aplasia after treatment with recombinant erythropoietin products: recommendations for minimization of risk. J Am Soc Nephrol 2004; 15:2728.
  20. Macdougall IC. Antibody-mediated pure red cell aplasia (PRCA): epidemiology, immunogenicity and risks. Nephrol Dial Transplant 2005; 20 Suppl 4:iv9.
  21. Macdougall IC, Casadevall N, Locatelli F, et al. Incidence of erythropoietin antibody-mediated pure red cell aplasia: the Prospective Immunogenicity Surveillance Registry (PRIMS). Nephrol Dial Transplant 2015; 30:451.
  22. Macdougall IC. Pure red cell aplasia with anti-erythropoietin antibodies occurs more commonly with one formulation of epoetin alfa than another. Curr Med Res Opin 2004; 20:83.
  23. Amgen Statement on Pure Red Cell Aplasia www.amgen.com/clinicians/prca.html (Accessed on March 07, 2005).
  24. Janssen Research and Development Case Reports www.jnjpharmarnd.com/company/n-casereports.html. (Accessed on December 13, 2005).
  25. Locatelli F, Del Vecchio L, Pozzoni P. Pure red-cell aplasia "epidemic"--mystery completely revealed? Perit Dial Int 2007; 27 Suppl 2:S303.
  26. McKoy JM, Stonecash RE, Cournoyer D, et al. Epoetin-associated pure red cell aplasia: past, present, and future considerations. Transfusion 2008; 48:1754.
  27. Alonso Melgar A, Melgosa Hijosa M, Pardo de la Vega R, et al. Antierythropoietin antibody-induced pure red cell aplasia: posttransplant evolution. Pediatr Nephrol 2004; 19:1059.
  28. Praditpornsilpa K, Tiranathanagul K, Kupatawintu P, et al. Biosimilar recombinant human erythropoietin induces the production of neutralizing antibodies. Kidney Int 2011; 80:88.
  29. Haag-Weber M, Eckardt KU, Hörl WH, et al. Safety, immunogenicity and efficacy of subcutaneous biosimilar epoetin-α (HX575) in non-dialysis patients with renal anemia: a multi-center, randomized, double-blind study. Clin Nephrol 2012; 77:8.
  30. Panichi V, Ricchiuti G, Scatena A, et al. Pure red cell aplasia induced by epoetin zeta. Clin Kidney J 2016; 9:599.
  31. Fishbane S, Singh B, Kumbhat S, et al. Intravenous Epoetin Alfa-epbx versus Epoetin Alfa for Treatment of Anemia in End-Stage Kidney Disease. Clin J Am Soc Nephrol 2018; 13:1204.
  32. Wu G, Wadgymar A, Wong G, et al. A cross-sectional immunosurveillance study of anti-EPO antibody levels in CRF patients receiving epoetin alfa in 5 Ontario Renal Centers. Am J Kidney Dis 2004; 44:264.
  33. Stoffel MP, Haverkamp H, Kromminga A, et al. Prevalence of anti-erythropoietin antibodies in hemodialysis patients without clinical signs of pure red cell aplasia. Comparison between hypo- and normoresponsive patients treated with epoetins for renal anemia. Nephron Clin Pract 2007; 105:c90.
  34. Chapter 1: Diagnosis and evaluation of anemia in CKD. Kidney Int Suppl (2011) 2012; 2:288.
  35. Casadevall N, Cournoyer D, Marsh J, et al. Recommendations on haematological criteria for the diagnosis of epoetin-induced pure red cell aplasia. Eur J Haematol 2004; 73:389.
  36. Weber G, Gross J, Kromminga A, et al. Allergic skin and systemic reactions in a patient with pure red cell aplasia and anti-erythropoietin antibodies challenged with different epoetins. J Am Soc Nephrol 2002; 13:2381.
  37. Pollock C, Johnson DW, Hörl WH, et al. Pure red cell aplasia induced by erythropoiesis-stimulating agents. Clin J Am Soc Nephrol 2008; 3:193.
  38. Swanson SJ, Ferbas J, Mayeux P, Casadevall N. Evaluation of methods to detect and characterize antibodies against recombinant human erythropoietin. Nephron Clin Pract 2004; 96:c88.
  39. Thorpe R, Swanson SJ. Assays for detecting and diagnosing antibody-mediated pure red cell aplasia (PRCA): an assessment of available procedures. Nephrol Dial Transplant 2005; 20 Suppl 4:iv16.
  40. Wadhwa M, Mytych DT, Bird C, et al. Establishment of the first WHO Erythropoietin antibody reference panel: Report of an international collaborative study. J Immunol Methods 2016; 435:32.
  41. Amgen, Inc., Medical Information. Direct communication, June 16, 2005.
  42. Asari A, Gokal R. Pure red cell aplasia secondary to epoetin alpha responding to Darbepoetin alpha in a patient on peritoneal dialysis. J Am Soc Nephrol 2004; 15:2204.
  43. Summers SA, Matijevic A, Almond MK. Successful re-introduction of recombinant human erythropoietin following antibody induced pure red cell aplasia. Nephrol Dial Transplant 2004; 19:2137.
  44. Macdougall IC, Roche A, Rossert J, et al. Re-challenging patients who developed pure red cell aplasia with epoetin: can it be done? Nephrol Dial Transplant 2004; 19:2901.
  45. Rossert J, Macdougall I, Casadevall N. Antibody-mediated pure red cell aplasia (PRCA) treatment and re-treatment: multiple options. Nephrol Dial Transplant 2005; 20 Suppl 4:iv23.
  46. Lim SK, Bee PC, Keng TC, Chong YB. Resolution of epoetin-induced pure red cell aplasia 2 years later, successful re-challenge with continuous erythropoiesis receptor stimulator. Clin Nephrol 2013; 80:227.
  47. Hirai K, Ookawara S, Miyazawa H, et al. Successful treatment of a hemodialyzed patient with pure red cell aplasia associated with epoetin beta pegol therapy with cyclosporine. CEN Case Rep 2016; 5:78.
  48. Viron B, Dupuy CA, Kolta A, Casadevall N. Successful re-challenge with darbepoetin in a patient with rHu-EPO-induced pure red cell aplasia refractory to immunosuppressive drugs. Nephrol Dial Transplant 2008; 23:2416.
  49. Verhelst D, Rossert J, Casadevall N, et al. Treatment of erythropoietin-induced pure red cell aplasia: a retrospective study. Lancet 2004; 363:1768.
  50. Chng WJ, Tan LK, Liu TC. Cyclosporine treatment for patients with CRF who developed pure red blood cell aplasia following EPO therapy. Am J Kidney Dis 2003; 41:692.
  51. Mandreoli M, Finelli C, Lopez A, et al. Successful resumption of epoetin alfa after rituximab treatment in a patient with pure red cell aplasia. Am J Kidney Dis 2004; 44:757.
  52. Comont T, Bournet B, Casadevall N, et al. Rituximab in pure red-cell aplasia secondary to anti-erythropoietin antibodies. Kidney Int 2014; 86:210.
  53. Hashimoto K, Harada M, Kamijo Y. Pure red cell aplasia induced by anti-erythropoietin antibodies, well-controlled with tacrolimus. Int J Hematol 2016; 104:502.
  54. Bennett CL, Cournoyer D, Carson KR, et al. Long-term outcome of individuals with pure red cell aplasia and antierythropoietin antibodies in patients treated with recombinant epoetin: a follow-up report from the Research on Adverse Drug Events and Reports (RADAR) Project. Blood 2005; 106:3343.
  55. Covic A, Cannata-Andia J, Cancarini G, et al. Biosimilars and biopharmaceuticals: what the nephrologists need to know--a position paper by the ERA-EDTA Council. Nephrol Dial Transplant 2008; 23:3731.
  56. Wu Y, Cai X, Ni J, Lin X. Resolution of epoetin-induced pure red cell aplasia, successful re-challenge with roxadustat. Int J Lab Hematol 2020; 42:e291.
  57. Wan K, Yin Y, Luo Z, Cheng J. Remarkable response to roxadustat in a case of anti-erythropoietin antibody-mediated pure red cell aplasia. Ann Hematol 2021; 100:591.
  58. Wu R, Peng Y. Roxadustat on anti-erythropoietin antibody-related pure red cell aplasia in the patient with end-stage renal disease. Semin Dial 2021; 34:319.
  59. Cai KD, Zhu BX, Lin HX, Luo Q. Successful application of roxadustat in the treatment of patients with anti-erythropoietin antibody-mediated renal anaemia: a case report and literature review. J Int Med Res 2021; 49:3000605211005984.
  60. Zhang H, Huang Z, He L, et al. Successful treatment of anti-EPO antibody associated refractory anemia with hypoxia-inducible factor prolyl hydroxylase inhibitor. Ren Fail 2020; 42:860.
Topic 1930 Version 34.0

References