Back To Search Results

Erythropoietin Stimulating Agents

Editor: Judith Borger Updated: 7/14/2024 9:04:03 PM

Indications

Endogenous erythropoietin (EPO) is a glycoprotein hormone that stimulates red blood cell production. The peritubular cells of the renal cortex produce most of the EPO found in a human adult, with the liver serving as the primary site of fetal production.[1] EPO production can also occur in small quantities in the spleen, liver, bone marrow, lung, and brain. A reduced partial pressure of oxygen (pO2) directly stimulates EPO production. Reduced hemoglobin levels also stimulate EPO production, albeit indirectly. 

Erythropoiesis stimulating agents (ESAs) are recombinant forms of EPO produced synthetically via recombinant DNA technology in cell cultures. Examples of erythropoiesis-stimulating agents include epoetin alfa, darbepoetin, and methoxy polyethylene glycol-epoetin β.[1]

ESAs are generally indicated for patients with conditions associated with impaired red blood cell production. The 2 primary FDA-approved indications for ESA administration are 1) anemia secondary to chronic kidney disease and 2) chemotherapy-induced anemia in patients with cancer.[2] The FDA approved the use of epoetin (1993) and darbepoetin (2002) for patients with chemotherapy-induced anemia. The FDA has also approved ESAs for the treatment of anemia secondary to zidovudine therapy for HIV infection, patients receiving autologous blood transfusions, patients with anemia undergoing elective surgery (pre-op and post-op), and anemia in preterm infants.

Erythropoiesis-stimulating agents (ESAs) have also demonstrated therapeutic benefits for patients with chronic kidney disease (CKD), a condition characterized by renal damage and reduced erythropoietin (EPO) production by peritubular cells.[3] ESAs are beneficial for patients receiving dialysis or who will soon need dialysis. In patients with CKD and chemotherapy-induced anemia, ESAs are generally reserved for patients with a hemoglobin <10 g/dL due to the risk of adverse effects.[2]

The recommendations of the American Society of Clinical Oncology/American Society of Hematology (ASCO/ASH) are summarized below.[4]

  • Depending on clinical circumstances, erythropoiesis-stimulating agents (ESAs) may be prescribed to patients with chemotherapy-associated anemia when cancer treatment is not expected to be curative and the hemoglobin is <10 g/dL.
  • ESAs should not be prescribed to patients with chemotherapy-associated anemia whose cancer treatment is expected to be curative. Additionally, ESAs should not be prescribed to most patients with nonchemotherapy-associated anemia. 
  • The American Society of Clinical Oncology (ASCO) considers epoetin, darbepoetin, and biosimilars equally effective and safe. Biosimilars of epoetin alfa are safe and effective compared to their progenitor. However, the evidence is primarily derived from studies in patients with cancer and chronic kidney disease, with moderate to low quality. Biosimilars have been used in Europe for over 10 years without significant concerns. Providers in the United States should review approvals and indications based on local regulatory authorities.
  • When prescribing ESAs, providers should aim to elevate hemoglobin to the lowest concentration necessary to avoid or reduce the need for red blood cell transfusions. If no response is observed within 6 to 8 weeks, ESA therapy should be discontinued, and the patient should be reevaluated for underlying causes, such as tumor progression or iron deficiency.[4]

FDA-Approved Indications

Epoetin alfa:

  • Anemia associated with chronic kidney disease
  • Anemia associated with HIV infection in patients receiving ≤ 4200 mg/week of zidovudine, with endogenous serum erythropoietin levels ≤ 500 units/mL
  • Simultaneous myelosuppressive chemotherapy requiring at least 2 additional months of planned chemotherapy
  • Reduction of allogeneic RBC transfusions in high-risk patients undergoing noncardiac, nonvascular (elective) surgery
  • Reduction of allogeneic RBC transfusions in high-risk patients with a perioperative hemoglobin of 10-13 g/dL

Darbepoetin:

  • Anemia associated with chronic kidney disease
  • Treatment of anemia in patients with non-myeloid malignancies currently undergoing myelosuppressive chemotherapy, with at least 2 additional months of chemotherapy planned upon initiation

Methoxy polyethylene glycol-epoetin β [pegylated continuous erythropoietin receptor activator (CERA)]

  • Anemia associated with chronic kidney disease, regardless of dialysis status
  • Anemia associated with CKD in children 3 months and older who are switching from another erythropoiesis-stimulating agent (ESA) after achieving stable hemoglobin levels. This indication is independent of dialysis status.[5]

Off-Label Uses

  • ASCO suggests that ESAs may also be considered for patients with lower-risk myelodysplastic syndromes and a serum erythropoietin level ≤500 IU/L.[4]
  • A systematic review and network meta-analysis of randomized controlled trials assessed therapeutic strategies for reducing red blood cell (RBC) transfusion in ICU patients. Combination therapy with iron and erythropoiesis-stimulating agents reduced transfusion requirement compared to placebo/no treatment (RR, 0.60 [95% CI, 0.49-0.74]). Iron and erythropoietin monotherapies also demonstrated potential benefits. However, given the risks associated with ESA therapy, this indication warrants further investigation.[6]

Mechanism of Action

Register For Free And Read The Full Article
Get the answers you need instantly with the StatPearls Clinical Decision Support tool. StatPearls spent the last decade developing the largest and most updated Point-of Care resource ever developed. Earn CME/CE by searching and reading articles.
  • Dropdown arrow Search engine and full access to all medical articles
  • Dropdown arrow 10 free questions in your specialty
  • Dropdown arrow Free CME/CE Activities
  • Dropdown arrow Free daily question in your email
  • Dropdown arrow Save favorite articles to your dashboard
  • Dropdown arrow Emails offering discounts

Learn more about a Subscription to StatPearls Point-of-Care

Mechanism of Action

Endogenous erythropoietin and erythropoiesis-stimulating agents induce the division and differentiation of erythroid progenitor cells.[3] The surface of CD34+ hematopoietic stem cells (erythrocyte progenitor cells) contains EPO receptors. The binding of endogenous EPO or recombinant analogs creates a cellular signaling cascade, activating genes that promote cell proliferation and prevent apoptosis. The result is an elevation in total body hemoglobin and hematocrit.

The continuous erythropoietin receptor activator (CERA) methoxy polyethylene glycol-epoetin β is a synthetic version of erythropoietin that releases epoetin β slowly. This medication has a longer half-life than other ESAs and may be administered on a monthly basis.[5] Severe pre-transfusion anemia in ICU patients enhances the hemoglobin increment post-transfusion. During studies observing mice, erythropoietin promoted RBC recovery post-transfusion, which is crucial for managing hypoxemia involving erythroid progenitors and macrophages.[7]

Pharmacokinetics

Absorption: Subcutaneous administration of epoetin alfa achieves peak plasma concentration (Cmax) within 5 to 24 hours. Darbepoetin has a bioavailability of approximately 37% in adults and 54% in children. Hemoglobin levels typically increase 2 to 6 weeks after starting treatment. The absolute bioavailability of methoxy polyethylene glycol-epoetin β after subcutaneous administration is 62%.

Distribution: The volume of distribution for epoetin alfa and methoxy polyethylene glycol-epoetin β is approximately 61 mL/kg. Darbepoetin is found in maximum concentrations in bone marrow or the injection site (skin), with subsequent distribution to the thyroid gland, kidneys, adrenal glands, spleen, lungs, stomach, and bladder.

Metabolism: Erythropoietin and epoetin alfa are internalized and degraded after binding to the erythropoietin receptor (EPO-R). ESAs may also be cleared via the reticuloendothelial and lymphatic systems. The removal of erythropoietin's oligosaccharide side chains primarily occurs in the liver. Methoxy polyethylene glycol–epoetin β has a structure similar to epoetin but is enhanced with a water-soluble polyethylene glycol group. Pegylation increases its molecular weight, enhancing its solubility and protecting it from enzymatic breakdown.

Excretion: Epoetin-alpha has a half-life of 6 hours after intravenous administration and 24 hours after subcutaneous administration.[8] The average terminal half-life of darbepoetin in adults after subcutaneous administration (t1/2 = 48.4 h) exceeds that of intravenous (t1/2 = 25.3 h). This phenomenon (termed "flip-flop kinetics") is observed when a drug's absorption is significantly slower than its elimination, causing its persistence in the body to be chiefly influenced by absorption rather than elimination.[9][10] Pegylated methoxy polyethylene glycol–epoetin β demonstrates reduced renal clearance. This structural modification extends its half-life to approximately 130 hours following subcutaneous or intravenous administration.[11] Importantly, half-life and other pharmacokinetic parameters vary based on factors such as chemotherapy, CKD, age, and route of administration.

Administration

Available Dosage Forms and Strengths 

  • During large-scale manufacturing protocols, erythropoiesis-stimulating agents are typically sourced from transfected Chinese hamster ovary cells (CHOs).[3] An isotonic solution buffers the ESA powder, which the provider can administer intravenously or subcutaneously. 
  • Darbepoetin is available in 25 μg, 40 μg, 60 μg, 100 μg, and 200 μg single-dose powder vials and premixed, single-dose syringes with the following powder and buffer dilutions: 10 μg in 0.4 mL, 25 μg in 0.42 mL, 40 μg in 0.4 mL, 60 μg in 0.3 mL, 100 μg in 0.5 mL, 300 μg in 0.6 mL, and 500 μg in 1 mL.
  • Epoetin alfa is available in 2,000 IU/mL, 3,000 IU/mL, 4,000 IU/mL, and 10,000 IU/mL single-dose vials. Additionally, it is available in 10,000 IU/mL and 20,000 IU/mL multiple-dose vials containing benzyl alcohol. 
  • Methoxy polyethylene glycol-epoetin β is available in 30 μg, 50 μg, 75 μg, 100 μg, 120 μg, 150 μg, 200 μg, or 250 μg in 0.3 mL solution in single-dose, prefilled, injectable syringes. A 360 μg in 0.6 mL solution is also available in a single-dose prefilled syringe for once-monthly administration.

Adult Dosing

Below are the standard dosing protocols for ESA administration. Dosing and frequency can be adjusted based on response to treatment. Specific and alternate protocols should be verified via manufacturer information or institutional protocols.

Epoetin alfa:

  • Chronic kidney disease-associated anemia: Start with 50 to 100 units/kg (IV or SC), 3 doses/week. IV dosing is preferred for patients on hemodialysis.
  • HIV-associated anemia: Start with 100 units/kg/dose (IV or SC), 3 doses/week. The maximum dosing is 300 units/kg/dose, 3 doses/week.
  • Chemotherapy-related anemia: Start with 40,000 units (SC) once weekly or 150 units/kg/dose (SC), 3 doses/week.
  • Surgery-associated transfusion reduction: 300 units/kg/dose (SC) daily for 15 days

Darbepoetin alfa:

  • Chronic kidney disease-associated anemia: Start with 0.45 μg/kg (IV or SC) once weekly.
  • Chemotherapy-related anemia: Start with 2.25 μg/kg/dose (SC) once weekly or 500 μg (SC) every 3 weeks.

Methoxy polyethylene glycol-epoetin β:

Before initiating this medication, ensure the patient's hemoglobin level is stable on current epoetin or darbepoetin dosing. The patient's iron status should be determined before and routinely during ESA therapy. Supplemental iron should be given if serum ferritin is <100 μg/L or transferrin saturation is <20%.

  • Patients not currently undergoing ESA therapy:
    • On dialysis: Start with 0.6 μg/kg once every 2 weeks.
    • Not on dialysis: Start with 1.2 μg/kg (SC) monthly. Alternatively, a 0.6 μg/kg (IV or SC) starting dose can be administered once every 2 weeks.
  • Conversion from another ESA:
    • Adjust dosing to once monthly or once every 2 weeks based on the total weekly dose of epoetin alfa or darbepoetin alfa at the time of conversion.

Specific Patient Populations

Hepatic impairment: The manufacturer's label for epoetin alfa, methoxy polyethylene glycol-epoetin β, and darbepoetin does not provide dosage adjustments. Metabolic Associated Fatty Liver Disease (MAFLD) and advanced liver fibrosis were not found to correlate with hypo-responsiveness to erythropoiesis-stimulating agents (ESAs) independently. However, the higher FIB-4(Fibrosis-4) score observed in the ESA hypo-responsive group and the association between liver stiffness measurement and ESA hypo-responsiveness suggests that liver fibrosis could be a potential clinical indicator of ESA hypo-responsiveness.[12]

Renal impairment: Epoetin alfa, methoxy polyethylene glycol-epoetin β, and darbepoetin are indicated for chronic kidney anemia, as mentioned above. A post-marketing study assessed long-term darbepoetin in Japanese patients with non-dialysis chronic kidney disease (CKD). Cardiovascular adverse events occurred in 12.6% of the study population. Higher hemoglobin (Hb) levels (≥11 g/dL) were associated with fewer composite renal endpoints without an increase in cardiovascular adverse events (p < 0.0001).[13] 

Pregnancy considerations: Epoetin alfa is the preferred ESA during pregnancy due to comprehensive safety data compared to other agents.[14] Epoetin alfa should be considered for severe anemia in iron-replete patients who are not responding adequately to intravenous iron alone. Multi-use vials containing benzyl alcohol must not be administered to women who are pregnant or lactating; single-use vials without benzyl alcohol are preferred. Darbepoetin alfa should be used cautiously in women who are pregnant and on dialysis due to limited safety data. Successful outcomes have been reported with weekly doses up to 100 μg, but risks and benefits must be carefully weighed. Methoxy polyethylene glycol has limited use for women who are pregnant, with few successful outcomes documented. Further research is needed to establish its safety profile compared to that of epoetin alfa.

Breastfeeding considerations: 

Erythropoietin is naturally present in human milk and may support mammary epithelial and infant gastrointestinal tract health, hypothetically reducing the risk of HIV transmission.[15][16]

Epoetin alfa has been determined safe for women who are breastfeeding. Some study data suggest improved postpartum anemia response when epoetin alfa is combined with iron therapy, but the current consensus indicates it does not clinically enhance hemoglobin levels over iron alone. No adverse effects have been reported in breastfed infants whose mothers received epoetin alfa. Due to the benzyl alcohol content, the manufacturer recommends avoiding multiple-dose vials during lactation and delaying breastfeeding for 2 weeks after any dose containing benzyl alcohol. No special precautions are needed for women who are breastfeeding and taking epoetin alfa from single-use vials without preservatives.

The excretion of darbepoetin alfa into breast milk and its effects on breastfed infants have not been specifically studied. Darbepoetin alfa is immunologically and biologically similar to native erythropoietin. Intravenous administration of darbepoetin alfa to newborn infants has been safely conducted using doses exceeding those anticipated to be excreted into breast milk. Therefore, no special precautions are necessary while breastfeeding.[17]

The excretion of methoxy polyethylene glycol-epoetin β into breast milk has not been studied. Due to the lack of specific data on methoxy polyethylene glycol-epoetin β in breastfeeding, an alternative medication may be preferable, particularly when nursing a newborn or preterm infant.[18]

Pediatric patients:

In a randomized multicenter study, preterm infants receiving darbepoetin or erythropoietin showed improved cognitive outcomes at 18 to 22 months compared to placebo. These ESAs were associated with higher cognitive scores and better cognitive permanence.[19]

Epoetin alfa is recommended for patients 1 month and older with CKD-associated anemia. However, its safety and effectiveness have not been established for patients younger than 1 month. Epoetin alfa is recommended for patients 5 and older with chemotherapy-related anemia, but its safety and efficacy have not been established for patients younger than 5.

Darbepoetin alfa is indicated for patients with CKD who are 1 month or older. However, its safety and effectiveness have not been established for patients younger than 1 month, and its use in pediatric cancer has not been established.

Methoxy polyethylene glycol-epoetin β appears to have a favorable safety profile.[20] This medication benefits patients with anemia who are 3 months or older after stabilization with another ESA. This benefit is independent of dialysis status. However, its safety and effectiveness have not been established in patients younger than 3 months. Continuous erythropoietin receptor activator (CERA)/methoxy polyethylene glycol-epoetin β is associated with stable hemoglobin levels in pediatric patients with chronic kidney disease (CKD) who are on dialysis.

Older patients: Careful consideration is necessary when selecting the ESA dose, and providers are encouraged to start at the lower end of the dosing spectrum. This approach acknowledges the higher incidence of compromised hepatic, renal, or cardiac function and the presence of concurrent illnesses or medications in older adults.

Adverse Effects

The most severe adverse effects of EPO are related to a significant risk of thrombotic events, particularly in surgical patients.[3] Supplemental use of erythropoiesis-stimulating agents increases blood viscosity. Given this, as well as the reduced vasodilatory effect due to a low baseline pO2, there is an associated increased risk of ischemic stroke and myocardial infarction.[21] There is also an increased risk of venous thromboembolism, and some have proposed the use of antithrombotic prophylaxis in patients receiving ESA therapy.[3]

There is also concern regarding the potential progression of tumorigenesis in patients with certain forms of cancer, particularly breast cancer, non-small cell lung cancer, head and neck cancer, lymphoid cancer, and cervical cancer.[22] This occurs through increased cell signaling and tumor angiogenesis.

In a multicenter study, nausea, vomiting, diarrhea, fatigue, insomnia, peripheral edema, thrombocytopenia, myalgias, arthralgias, rashes, abdominal pain, headache, and paresthesias were common adverse effects experienced by patients undergoing chemotherapy and epoetin alfa treatment.[23]

Drug-Drug Interactions

Hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF-PHIs): Due to synergism, concurrent administration of ESA with HIF-PHIs, such as roxadustat and vadadustat, is not advisable.

ACE inhibitors: In a study of 660 hemodialysis patients, ACE inhibitor therapy was associated with lower hemoglobin levels (p=0.02) and higher erythropoietin resistance in those with the ACE gene D/D genotype (p=0.006). Additional research is required.[24]

Angiotensin receptor blockers: In one study, researchers explored the effects of angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs) on erythropoiesis in hemodialysis patients and healthy controls. They observed that ARBs significantly reduced the formation of erythroid progenitor colonies (burst-forming units-erythroid) in both groups, whereas ACE inhibitors had a lesser impact. This suggests that ARB-mediated AT1 receptor blockade directly inhibits erythropoiesis in vitro. Further research is required.[25]

Contraindications

Epoetin alfa, darbepoetin, and methoxy polyethylene glycol-epoetin β are contraindicated for patients with uncontrolled hypertension, pure red cell aplasia (PRCA), and severe allergic reactions. Erythropoietin-stimulating agents are contraindicated for patients with hypersensitivity to non-human mammal-derived products because of ESA production methods.[3] ESAs containing benzyl alcohol are contraindicated for neonates, peripartum mothers, and breastfeeding mothers due to the risk of gasping syndrome. This syndrome causes gasping respiration, renal failure, and neurological deterioration in neonates, resulting from severe metabolic acidosis.[26]

Box Warnings

ESAs increase mortality, myocardial infarction, thrombosis of vascular access, stroke, venous thromboembolism, and tumor progression or recurrence in certain patient populations.[27][28]

Chronic kidney disease: Patients who were given erythropoiesis-stimulating agents (ESAs) to achieve a hemoglobin level above 11 g/dL were at a higher risk of death, serious adverse cardiovascular events, and stroke. Providers should administer the lowest ESA dose sufficient to reduce the need for red blood cell transfusions.

Cancer: ESAs are associated with decreased overall survival and increased risk of tumor progression/recurrence in patients with non-small cell lung, head and neck, lymphoid, cervical, and breast cancers. Providers should administer the lowest effective ESA dose to decrease the risk of RBC transfusions and serious cardiovascular and thromboembolic reactions. ESAs should only be used to manage anemia caused by myelosuppressive chemotherapy. They should not be prescribed for patients undergoing myelosuppressive chemotherapy with the intention of cure. ESAs are discontinued following the completion of a chemotherapy course.

Warning and Precautions

Thromboembolic events: The American College of Cardiology/American Heart Association (ACC/AHA) 2022 guidelines note that anemia in heart failure (HF) is associated with impaired erythropoietin production and is linked to worse long-term outcomes. Although initial small study data suggested the potential benefits of erythropoiesis-stimulating agents (ESAs) for improving functional capacity and reducing hospitalizations in patients with HF, there is a risk of thromboembolic events, including stroke.[29] In patients with HF and anemia, ESAs should not be prescribed to improve morbidity and mortality.[30] Physicians should exercise caution when prescribing ESAs to patients with a history of deep vein thrombosis (DVT), pulmonary embolism, or hypercoagulability disorder. Similar caution is warranted for patients with a history of ischemic stroke or cardiovascular disease, owing to the potential increase in blood viscosity associated with ESA use.[21]

Pure red cell aplasia (PRCA): Characterized by severe anemia and low reticulocyte count, PRCA has been reported in postmarketing to be associated with neutralizing antibodies against erythropoietin, along with methoxy polyethylene glycol-epoetin β, darbepoetin, and epoetin. This risk is prominent in patients with CKD who are administered ESAs subcutaneously. PRCA has also occurred in patients receiving ESAs for hepatitis C-related anemia, a use that the FDA has not approved.[31][32] PRCA has been observed in patients receiving darbepoetin alfa and epoetin alfa therapy. ESA therapy should be discontinued in patients who develop severe anemia and low reticulocyte count during treatment, and the presence of erythropoietin-neutralizing antibodies should be determined. ESAs should be discontinued permanently if PRCA develops. Switching to another ESA is not advisable as it may lead to cross-reactivity due to antibody formation.

Seizures: ESAs increase the risk of seizures in patients with CKD. These patients should be monitored closely for neurologic symptoms during treatment initiation. New-onset seizures, premonitory symptoms, or altered seizure frequency should prompt emergency medical attention.[33]

Allergic reactions: Severe allergic reactions, including anaphylactic reactions, angioedema, bronchospasm, skin rash, and urticaria, have been reported in patients receiving methoxy polyethylene glycol–epoetin β, darbepoetin alfa, and epoetin alfa. Patients should be monitored closely for clinical signs and symptoms. Providers should immediately discontinue treatment and administer appropriate therapy if such reactions occur.

Severe cutaneous adverse drug reactions: Stevens-Johnson syndrome, erythema multiforme, and toxic epidermal necrolysis have been reported in patients treated with erythropoiesis-stimulating agents (ESAs).[34]

Potential for abuse: Although ESAs are not controlled substances, they may be used by athletes for performance-enhancing purposes.[35]

Dialysis monitoring:  The initiation of ESAs may require an increased dosage of heparin for anticoagulation to prevent clotting of the extracorporeal circuit during hemodialysis. One study identified higher doses of ESAs and doxazosin as risk factors requiring further research.[36]

Monitoring

Patients receiving erythropoietin-stimulating agent therapy should have baseline hemoglobin and transferrin levels documented before initiating.[3] Hemoglobin should be checked weekly after beginning treatment. The dosing and administration frequency should be adjusted based on the response to treatment. Providers should withhold treatment if hemoglobin rises to a non-anemic level.[37] Iron supplementation should be considered in patients with poor response to ESA therapy, as iron availability may be inadequate.[3] According to the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines, ESAs increase iron utilization and decrease serum iron, TSAT, and ferritin levels. They also suppress hepcidin by inducing erythropoiesis, increasing iron supply from macrophage stores and dietary sources. ESA stimulation can lead to or exacerbate iron deficiency by creating a high demand for iron that exceeds available stores. This may occur despite adequate initial iron levels, especially during inflammation, which elevates hepcidin and restricts iron release. Therefore, the patient's response to ESA therapy depends on iron status and inflammatory severity. Monitoring reticulocyte Hb content within 3–4 days of iron therapy initiation provides information regarding effective iron incorporation into red blood cells, aiding in tailoring iron and ESA therapy.

According to ASCO/ASH guidelines, for patients with non-Hodgkin lymphoma, myeloma, or chronic lymphocytic leukemia, clinicians should monitor the hematologic response to cancer treatment before considering an ESA. Caution is advised when using ESAs concomitantly with treatments/diseases that increase the risk of thromboembolic complications. Before initiating ESA therapy, healthcare providers should conduct a comprehensive assessment, including a review of the patient's medical history, physical examination, and diagnostic tests to identify and treat other potential causes of anemia apart from chemotherapy or underlying blood-related cancers. This assessment may involve examining a peripheral blood smear, conducting iron studies (including iron levels, total iron-binding capacity, transferrin saturation, and ferritin), checking for folate and vitamin B12 levels, assessing the reticulocyte count, looking for signs of hidden blood loss, evaluating renal function, measuring baseline erythropoietin levels, and checking serum thyroid-stimulating hormone levels when necessary. Additionally, some patients may require direct antiglobulin testing, such as the Coombs test.[4]

Vitamin B6 deficiency should be considered in pediatric patients on hemodialysis who are not responding to ESA and iron therapy. Supplemental vitamin B6-corrected anemia can improve hemoglobin levels without further transfusions.[38]

One study suggests that erythrocyte creatine content could be a promising predictor of anemia improvement in maintenance hemodialysis patients receiving erythropoiesis-stimulating agents (ESAs). However, further research is needed to validate its clinical utility and optimize its application in practice.[39]

Toxicity

Signs and Symptoms of Overdose

Most erythropoietin-stimulating agent toxicity cases are related to the adverse effects of chronic use. There are few case reports of acute toxicity. One involved a man who intentionally injected himself with recombinant human erythropoietin, causing his hemoglobin to rise to a dangerously high level.[40] Cases of severe hypertension have been reported following ESA overdosage. Another study linked Environmental Protection Agency (EPA) water supply data with patient records from the United States Renal Data System (USRDS), revealing that even low levels of lead in community water systems are associated with decreased hemoglobin levels and increased use of erythropoiesis-stimulating agents (ESAs) in patients with end-stage kidney disease (ESKD).[41]

Management of Overdose

Patients should receive supportive care for hypertension, IV fluids, and serial phlebotomy. Severe cases may require erythrocytapheresis.[42]

Enhancing Healthcare Team Outcomes

Treating anemia in patients using erythropoietin-stimulating agents (ESA) requires an interprofessional team of medical providers. This team should include a nephrologist for patients with chronic kidney disease, a hematologist/oncologist overseeing chemotherapy, appropriate nursing staff, a pharmacist for ESA dosing, and a phlebotomy or laboratory technician for blood sampling to monitor for improvement.

The healthcare team should arrange appropriate follow-up appointments to reassess the patient's health status and any adverse effects of the ESA; this is where specialty-trained nurses and pharmacists can play a significant role. Nurses will have the most frequent contact with the patient while providing education, monitoring for adverse effects, and administering the medication. Pharmacists with specialty oncology certification should monitor dosing, interactions, and lab values that may alter or stop the dosing of the ESA. Both nurses and pharmacists must chart and inform the treating physician of any findings, changes, or concerns.

Patients should receive counseling on the potential adverse effects and when to seek immediate medical care. Emergency medical providers should be aware of the adverse effects of ESAs to risk-stratify patients for venous thromboembolism, acute coronary syndrome, ischemic stroke, and other emergent conditions related to ESA.

Only through the coordinated effort of an interprofessional healthcare team can ESA therapy be administered safely and effectively, with minimal adverse events and optimal patient results.

Monitoring the hemoglobin level to optimize patient outcomes and minimize adverse effects is very important. A debate exists regarding the appropriate target hemoglobin in individuals treated with ESA. A pooled analysis of 9 randomized control trials on patients with chronic kidney disease indicates that patients have higher mortality and morbidity from cardiovascular-related events when the patient's hemoglobin falls below 10 g/dL.[43] This pooled analysis showed no survival benefit for patients treated with ESA when the hemoglobin exceeded 13 g/dL. However, there was a higher incidence of adverse events such as hypertension, vascular access thrombosis, and stroke. Current guidelines advocate for tailoring target hemoglobin levels to individual needs. Optimal levels typically range between 11 and 12 g/dL, considering that higher doses of ESAs can increase the risk of thrombotic events. An interprofessional team approach and communication among clinicians (MDs, DOs, NPs, PAs), hematologists, pharmacists, and nurses are crucial to decreasing potential adverse effects and improving patient outcomes related to erythropoiesis-stimulating agents.

References


[1]

Jelkmann W. Erythropoietin. Frontiers of hormone research. 2016:47():115-27. doi: 10.1159/000445174. Epub 2016 Jun 27     [PubMed PMID: 27348128]


[2]

Noxon V, Knopf KB, Norris LB, Chen B, Yang YT, Qureshi ZP, Hrushesky W, Lebby AA, Schooley B, Hikmet N, Dickson M, Thamer M, Cotter D, Yarnold PR, Bennett CL. Tale of Two Erythropoiesis-Stimulating Agents: Utilization, Dosing, Litigation, and Costs of Darbepoetin and Epoetin Among South Carolina Medicaid-Covered Patients With Cancer and Chemotherapy-Induced Anemia. Journal of oncology practice. 2017 Jun:13(6):e562-e573. doi: 10.1200/JOP.2016.019364. Epub 2017 May 15     [PubMed PMID: 28504901]


[3]

Jelkmann W. Physiology and pharmacology of erythropoietin. Transfusion medicine and hemotherapy : offizielles Organ der Deutschen Gesellschaft fur Transfusionsmedizin und Immunhamatologie. 2013 Oct:40(5):302-9. doi: 10.1159/000356193. Epub 2013 Jul 19     [PubMed PMID: 24273483]


[4]

Bohlius J, Bohlke K, Castelli R, Djulbegovic B, Lustberg MB, Martino M, Mountzios G, Peswani N, Porter L, Tanaka TN, Trifirò G, Yang H, Lazo-Langner A. Management of Cancer-Associated Anemia With Erythropoiesis-Stimulating Agents: ASCO/ASH Clinical Practice Guideline Update. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2019 May 20:37(15):1336-1351. doi: 10.1200/JCO.18.02142. Epub 2019 Apr 10     [PubMed PMID: 30969847]

Level 1 (high-level) evidence

[5]

Fischbach M, Wühl E, Reigner SCM, Morgan Z, Schaefer F. Efficacy and Long-Term Safety of C.E.R.A. Maintenance in Pediatric Hemodialysis Patients with Anemia of CKD. Clinical journal of the American Society of Nephrology : CJASN. 2018 Jan 6:13(1):81-90. doi: 10.2215/CJN.03570417. Epub 2017 Nov 2     [PubMed PMID: 29097481]


[6]

Yoshihiro S, Hongo T, Yamamoto M, Taito S, Kataoka Y. Pharmacotherapy for Reducing RBC Transfusion for Patients in the ICU: A Systematic Review and Network Meta-Analysis. Critical care medicine. 2024 Apr 1:52(4):618-625. doi: 10.1097/CCM.0000000000006114. Epub 2023 Nov 13     [PubMed PMID: 37962159]

Level 1 (high-level) evidence

[7]

Casimir M, Colard M, Dussiot M, Roussel C, Martinez A, Peyssonnaux C, Mayeux P, Benghiat S, Manceau S, Francois A, Marin N, Pène F, Buffet PA, Hermine O, Amireault P. Erythropoietin downregulates red blood cell clearance, increasing transfusion efficacy in severely anemic recipients. American journal of hematology. 2023 Dec:98(12):1923-1933. doi: 10.1002/ajh.27117. Epub 2023 Oct 4     [PubMed PMID: 37792521]


[8]

Czock D, Keller F. Clinical Pharmacokinetics and Pharmacodynamics of Roxadustat. Clinical pharmacokinetics. 2022 Mar:61(3):347-362. doi: 10.1007/s40262-021-01095-x. Epub 2021 Dec 14     [PubMed PMID: 34905154]


[9]

Kim S, Hong T, Ko JW, Huh W, Kim JR. Comparison of the Pharmacokinetic-Pharmacodynamic Relationships of Two Darbepoetin Alfa Formulations in Healthy Male Volunteers. BioDrugs : clinical immunotherapeutics, biopharmaceuticals and gene therapy. 2019 Feb:33(1):101-112. doi: 10.1007/s40259-018-0323-0. Epub     [PubMed PMID: 30506495]


[10]

An G, Ohls RK, Christensen RD, Widness JA, Mock DM, Veng-Pedersen P. Population Pharmacokinetics of Darbepoetin in Infants Following Single Intravenous and Subcutaneous Dosing. Journal of pharmaceutical sciences. 2017 Jun:106(6):1644-1649. doi: 10.1016/j.xphs.2017.02.001. Epub 2017 Feb 9     [PubMed PMID: 28189627]


[11]

Bantounou MA, Lamb A, Young D, Ramage IJ, Reynolds BC. Clinical Experience of a Long-acting Pegylated Erythropoietin-Stimulating Agent in Pediatric Chronic Kidney Disease. The journal of pediatric pharmacology and therapeutics : JPPT : the official journal of PPAG. 2023:28(6):509-518. doi: 10.5863/1551-6776-28.6.509. Epub 2023 Oct 28     [PubMed PMID: 38130347]


[12]

Wong WK, Chan WK, Ganapathy S, Lim SK. Is metabolic-dysfunction-associated fatty liver disease or advanced liver fibrosis associated with erythropoietin stimulating agent hypo-responsiveness among patients with end-stage kidney disease on haemodialysis? Nephrology (Carlton, Vic.). 2023 Aug:28(8):425-433. doi: 10.1111/nep.14186. Epub 2023 Jun 3     [PubMed PMID: 37269220]


[13]

Tanaka T, Nangaku M, Imai E, Tsubakihara Y, Kamai M, Wada M, Asada S, Akizawa T. Safety and effectiveness of long-term use of darbepoetin alfa in non-dialysis patients with chronic kidney disease: a post-marketing surveillance study in Japan. Clinical and experimental nephrology. 2019 Feb:23(2):231-243. doi: 10.1007/s10157-018-1632-9. Epub 2018 Sep 4     [PubMed PMID: 30182223]


[14]

Drambarean B, Mastalerz J, Wendt L, Toth-Manikowski S. Pharmacotherapy considerations in pregnant patients on hemodialysis. Hemodialysis international. International Symposium on Home Hemodialysis. 2023 Jul:27(3):212-223. doi: 10.1111/hdi.13107. Epub 2023 Jun 21     [PubMed PMID: 37345253]


[15]

Miller M, Iliff P, Stoltzfus RJ, Humphrey J. Breastmilk erythropoietin and mother-to-child HIV transmission through breastmilk. Lancet (London, England). 2002 Oct 19:360(9341):1246-8     [PubMed PMID: 12401271]


[16]

Epoetin Alfa,,, 2006     [PubMed PMID: 30000431]


[17]

. Darbepoetin. Drugs and Lactation Database (LactMed®). 2006:():     [PubMed PMID: 30000807]


[18]

. Epoetin Beta. Drugs and Lactation Database (LactMed®). 2006:():     [PubMed PMID: 30277694]


[19]

Ohls RK, Kamath-Rayne BD, Christensen RD, Wiedmeier SE, Rosenberg A, Fuller J, Lacy CB, Roohi M, Lambert DK, Burnett JJ, Pruckler B, Peceny H, Cannon DC, Lowe JR. Cognitive outcomes of preterm infants randomized to darbepoetin, erythropoietin, or placebo. Pediatrics. 2014 Jun:133(6):1023-30. doi: 10.1542/peds.2013-4307. Epub 2014 May 12     [PubMed PMID: 24819566]

Level 1 (high-level) evidence

[20]

Kohlhas L, Studer M, Rutten-Jacobs L, Reigner SM, Sander A, Yap HK, Vondrak K, Coccia PA, Cano F, Schmitt CP, Warady BA, Schaefer F, IPDN collaborators. Real-world evidence on the dosing and safety of C.E.R.A. in pediatric dialysis patients: findings from the International Pediatric Dialysis Network registries. Pediatric nephrology (Berlin, Germany). 2024 Mar:39(3):807-818. doi: 10.1007/s00467-023-05977-z. Epub 2023 Aug 11     [PubMed PMID: 37566114]


[21]

McCullough PA, Barnhart HX, Inrig JK, Reddan D, Sapp S, Patel UD, Singh AK, Szczech LA, Califf RM. Cardiovascular toxicity of epoetin-alfa in patients with chronic kidney disease. American journal of nephrology. 2013:37(6):549-58. doi: 10.1159/000351175. Epub 2013 May 25     [PubMed PMID: 23735819]

Level 1 (high-level) evidence

[22]

Annese T, Tamma R, Ruggieri S, Ribatti D. Erythropoietin in tumor angiogenesis. Experimental cell research. 2019 Jan 15:374(2):266-273. doi: 10.1016/j.yexcr.2018.12.013. Epub 2018 Dec 18     [PubMed PMID: 30576679]


[23]

Glaspy JA, Charu V, Luo D, Moyo V, Kamin M, Wilhelm FE. Initiation of epoetin-alpha therapy at a starting dose of 120,000 units once every 3 weeks in patients with cancer receiving chemotherapy: an open-label, multicenter study with randomized and nonrandomized treatment arms. Cancer. 2009 Mar 1:115(5):1121-31. doi: 10.1002/cncr.24127. Epub     [PubMed PMID: 19170225]

Level 1 (high-level) evidence

[24]

Kiss Z, Ambrus C, Kulcsár I, Szegedi J, Kiss I, ACEGENE-BB_HU workgroup, ACEGENE-BB HU workgroup. Effect of angiotensin-converting enzyme gene insertion/deletion polymorphism and angiotensin-converting enzyme inhibition on erythropoiesis in patients on haemodialysis. Journal of the renin-angiotensin-aldosterone system : JRAAS. 2015 Dec:16(4):1021-7. doi: 10.1177/1470320314535276. Epub 2014 Jul 7     [PubMed PMID: 25002133]


[25]

Naito M, Kawashima A, Akiba T, Takanashi M, Nihei H. Effects of an angiotensin II receptor antagonist and angiotensin-converting enzyme inhibitors on burst forming units-erythroid in chronic hemodialysis patients. American journal of nephrology. 2003 Sep-Oct:23(5):287-93     [PubMed PMID: 12897465]


[26]

Gershanik J, Boecler B, Ensley H, McCloskey S, George W. The gasping syndrome and benzyl alcohol poisoning. The New England journal of medicine. 1982 Nov 25:307(22):1384-8     [PubMed PMID: 7133084]


[27]

Manns BJ, Tonelli M. The new FDA labeling for ESA--implications for patients and providers. Clinical journal of the American Society of Nephrology : CJASN. 2012 Feb:7(2):348-53. doi: 10.2215/CJN.09960911. Epub 2012 Jan 19     [PubMed PMID: 22266575]


[28]

Vu K, Zhou J, Everhart A, Desai N, Herrin J, Jena AB, Ross JS, Shah ND, Karaca-Mandic P. Uptake of evidence by physicians: De-adoption of erythropoiesis-stimulating agents after the TREAT trial. BMC nephrology. 2021 Aug 21:22(1):284. doi: 10.1186/s12882-021-02491-y. Epub 2021 Aug 21     [PubMed PMID: 34419007]


[29]

Kang J, Park J, Lee JM, Park JJ, Choi DJ. The effects of erythropoiesis stimulating therapy for anemia in chronic heart failure: A meta-analysis of randomized clinical trials. International journal of cardiology. 2016 Sep 1:218():12-22. doi: 10.1016/j.ijcard.2016.04.187. Epub 2016 May 11     [PubMed PMID: 27209352]

Level 1 (high-level) evidence

[30]

Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, Stevenson LW, Vardeny O, Vest AR, Yancy CW. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Journal of the American College of Cardiology. 2022 May 3:79(17):e263-e421. doi: 10.1016/j.jacc.2021.12.012. Epub 2022 Apr 1     [PubMed PMID: 35379503]

Level 1 (high-level) evidence

[31]

Shingu Y, Nakata T, Sawai S, Tanaka H, Asai O, Tamagaki K, Nakatani K. 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 nephrology. 2020 Jul 14:21(1):275. doi: 10.1186/s12882-020-01934-2. Epub 2020 Jul 14     [PubMed PMID: 32664894]

Level 3 (low-level) evidence

[32]

Ghany MG, Strader DB, Thomas DL, Seeff LB, American Association for the Study of Liver Diseases. Diagnosis, management, and treatment of hepatitis C: an update. Hepatology (Baltimore, Md.). 2009 Apr:49(4):1335-74. doi: 10.1002/hep.22759. Epub     [PubMed PMID: 19330875]


[33]

Lim J, Yu CJ, Yu H, Ha SJ. Erythropoietin therapy improves endothelial function in patients with non-dialysis chronic kidney disease and anemia (EARNEST-CKD): A clinical study. Medicine. 2021 Oct 22:100(42):e27601. doi: 10.1097/MD.0000000000027601. Epub     [PubMed PMID: 34678911]


[34]

Gillis NK, Hicks JK, Bell GC, Daly AJ, Kanetsky PA, McLeod HL. Incidence and Triggers of Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis in a Large Cancer Patient Cohort. The Journal of investigative dermatology. 2017 Sep:137(9):2021-2023. doi: 10.1016/j.jid.2017.05.010. Epub 2017 May 23     [PubMed PMID: 28549953]


[35]

Rocca A, Martin L, Kuuranne T, Ericsson M, Marchand A, Leuenberger N. A fast screening method for the detection of CERA in dried blood spots. Drug testing and analysis. 2022 May:14(5):820-825. doi: 10.1002/dta.3142. Epub 2021 Aug 21     [PubMed PMID: 34380180]


[36]

Wärme A, Hadimeri H, Nasic S, Stegmayr B. The association of erythropoietin-stimulating agents and increased risk for AV-fistula dysfunction in hemodialysis patients. A retrospective analysis. BMC nephrology. 2021 Jan 18:22(1):30. doi: 10.1186/s12882-020-02209-6. Epub 2021 Jan 18     [PubMed PMID: 33461526]

Level 2 (mid-level) evidence

[37]

Koulouridis I, Alfayez M, Trikalinos TA, Balk EM, Jaber BL. Dose of erythropoiesis-stimulating agents and adverse outcomes in CKD: a metaregression analysis. American journal of kidney diseases : the official journal of the National Kidney Foundation. 2013 Jan:61(1):44-56. doi: 10.1053/j.ajkd.2012.07.014. Epub 2012 Aug 22     [PubMed PMID: 22921639]

Level 1 (high-level) evidence

[38]

Searcy K, Rainwater S, Jeroudi M, Baliga R. Erythropoietin-stimulating agent-resistant vitamin B(6) deficiency anemia in a pediatric patient on hemodialysis. Pediatric nephrology (Berlin, Germany). 2021 Feb:36(2):473-476. doi: 10.1007/s00467-020-04810-1. Epub 2020 Nov 6     [PubMed PMID: 33156411]


[39]

Hayashi O, Nakamura S, Sugiura T, Hasegawa S, Tsuka Y, Takahashi N, Kikuchi S, Matsumura K, Okumiya T, Baden M, Shiojima I. Diagnostic accuracy and clinical usefulness of erythrocyte creatine content to predict the improvement of anaemia in patients receiving maintenance haemodialysis. BMC nephrology. 2023 Jan 3:24(1):1. doi: 10.1186/s12882-022-03055-4. Epub 2023 Jan 3     [PubMed PMID: 36597041]


[40]

Brown KR, Carter W Jr, Lombardi GE. Recombinant erythropoietin overdose. The American journal of emergency medicine. 1993 Nov:11(6):619-21     [PubMed PMID: 8240567]

Level 3 (low-level) evidence

[41]

Danziger J, Mukamal KJ, Weinhandl E. Associations of Community Water Lead Concentrations with Hemoglobin Concentrations and Erythropoietin-Stimulating Agent Use among Patients with Advanced CKD. Journal of the American Society of Nephrology : JASN. 2021 Oct:32(10):2425-2434. doi: 10.1681/ASN.2020091281. Epub 2021 Jul 15     [PubMed PMID: 34266982]


[42]

Hoffman RS,Cobrin G,Nelson LS,Howland MA, Erythropoietin overdose treated with emergent erythropheresis. Veterinary and human toxicology. 2002 Jun;     [PubMed PMID: 12046968]

Level 3 (low-level) evidence

[43]

Locatelli F, de Francisco A, Deray G, Fliser D, Armstrong G, Dougherty FC, Ehrhard P. Mortality and cardiovascular morbidity associated with haemoglobin levels: a pooled analysis of randomised controlled trials. Nephron. Clinical practice. 2014:128(3-4):323-32. doi: 10.1159/000366478. Epub 2014 Dec 5     [PubMed PMID: 25503585]

Level 1 (high-level) evidence