Local Coverage Determination (LCD)

Erythropoiesis Stimulating Agents

L39237

Expand All | Collapse All
Proposed LCD
Proposed LCDs are works in progress that are available on the Medicare Coverage Database site for public review. Proposed LCDs are not necessarily a reflection of the current policies or practices of the contractor.

Document Note

Note History

Contractor Information

LCD Information

Document Information

Source LCD ID
N/A
LCD ID
L39237
Original ICD-9 LCD ID
Not Applicable
LCD Title
Erythropoiesis Stimulating Agents
Proposed LCD in Comment Period
N/A
Source Proposed LCD
DL39237
Original Effective Date
For services performed on or after 07/24/2022
Revision Effective Date
For services performed on or after 01/18/2024
Revision Ending Date
N/A
Retirement Date
N/A
Notice Period Start Date
06/09/2022
Notice Period End Date
07/23/2022
AMA CPT / ADA CDT / AHA NUBC Copyright Statement

CPT codes, descriptions and other data only are copyright 2023 American Medical Association. All Rights Reserved. Applicable FARS/HHSARS apply.

Fee schedules, relative value units, conversion factors and/or related components are not assigned by the AMA, are not part of CPT, and the AMA is not recommending their use. The AMA does not directly or indirectly practice medicine or dispense medical services. The AMA assumes no liability for data contained or not contained herein.

Current Dental Terminology © 2023 American Dental Association. All rights reserved.

Copyright © 2023, the American Hospital Association, Chicago, Illinois. Reproduced with permission. No portion of the American Hospital Association (AHA) copyrighted materials contained within this publication may be copied without the express written consent of the AHA. AHA copyrighted materials including the UB‐04 codes and descriptions may not be removed, copied, or utilized within any software, product, service, solution or derivative work without the written consent of the AHA. If an entity wishes to utilize any AHA materials, please contact the AHA at 312‐893‐6816.

Making copies or utilizing the content of the UB‐04 Manual, including the codes and/or descriptions, for internal purposes, resale and/or to be used in any product or publication; creating any modified or derivative work of the UB‐04 Manual and/or codes and descriptions; and/or making any commercial use of UB‐04 Manual or any portion thereof, including the codes and/or descriptions, is only authorized with an express license from the American Hospital Association. The American Hospital Association (the "AHA") has not reviewed, and is not responsible for, the completeness or accuracy of any information contained in this material, nor was the AHA or any of its affiliates, involved in the preparation of this material, or the analysis of information provided in the material. The views and/or positions presented in the material do not necessarily represent the views of the AHA. CMS and its products and services are not endorsed by the AHA or any of its affiliates.

Issue

Issue Description

This LCD outlines limited coverage for this service with specific details under Coverage Indications, Limitations and/or Medical Necessity.

Issue - Explanation of Change Between Proposed LCD and Final LCD

CMS National Coverage Policy

This LCD supplements but does not replace, modify or supersede existing Medicare applicable National Coverage Determinations (NCDs) or payment policy rules and regulations for Erythropoiesis Stimulating Agents. All providers who report services for Medicare payment must fully understand and follow all existing laws, regulations, and rules for Medicare payment for Erythropoiesis Stimulating Agents and must properly submit only valid claims for them.

Title XVIII of the Social Security Act, §1862(a)(1)(A) excludes expenses incurred for items or services which are not reasonable and necessary for the diagnosis or treatment of illness or injury or to improve the functioning of a malformed body member.

Title XVIII of the Social Security Act, §1881(b)(1) allows payment for services furnished to individuals who have been determined to have end stage renal disease.

CMS Internet-Only Manual, Pub. 100-02, Medicare Benefit Policy Manual, Chapter 1, §30 Drugs and Biologicals

CMS Internet-Only Manual, Pub. 100-02, Medicare Benefit Policy Manual, Chapter 13, §120 Services and Supplies Furnished “Incident to” Physician’s Services

CMS Internet-Only Manual, Pub. 100-02, Medicare Benefit Policy Manual, Chapter 15, §50 Drugs and Biologicals, §50.1 Definition of Drug or Biological, §50.2 Determining Self-Administration of Drug or Biological, §50.3 Incident-to Requirements, §50.4.1 Approved Use of Drug, §50.4.3 Examples of Not Reasonable and Necessary, §50.5.2 Erythropoietin (EPO), §50.5.2.1 Requirements for Medicare Coverage for EPO, and §50.5.2.2 Medicare Coverage of Epoetin Alfa (Procrit) for Preoperative Use

CMS Internet-Only Manual, Pub. 100-03, Medicare National Coverage Determinations (NCD) Manual, Chapter 1, Part 2, §110.21 Erythropoiesis Stimulating Agents (ESAs) in Cancer and Related Neoplastic Conditions

NCD for ESAs for Cancer and Related Neoplastic Conditions - CAG-00383N (cms.gov)

National Coverage Analysis (NCA) for Erythropoiesis Stimulating Agents (ESAs) for Treatment of Anemia in Adults with CKD Including Patients on Dialysis and Patients not on Dialysis (CAG-00413N) (cms.gov)

CR 11244 Discontinuing the Erythropoietin Stimulating Agent (ESA) Monitoring Policy System Edits under the End Stage Renal Dialysis Prospective Payment System (ESRD PPS) Effective 01/01/2020 Pub 100-20 - One-Time Notification (PDF) (cms.gov)

Coverage Guidance

Coverage Indications, Limitations, and/or Medical Necessity

Erythropoietin (EPO) is naturally produced by the kidneys and stimulates the proliferation of red blood cells (RBCs) in the bone marrow. An erythropoietin stimulating agent (ESA) is a biologically engineered analog of EPO. ESAs contain the identical (or very similar) amino acid sequence as naturally occurring EPO and have the same biological effect. Several chronic conditions, especially chronic renal failure, result in decreased production of or relative resistance to EPO, often causing anemia. Supplementation by synthetic drugs with structures identical or similar to naturally occurring EPO has been proven safe and effective in correcting anemia in certain groups of patients. By elevating or maintaining the RBC level (as demonstrated by the hematocrit (HCT) and/or hemoglobin (Hb) levels), these synthetic analogs can decrease anemia and the need for transfusions.

Since 2007, the Food and Drug Administration (FDA) has issued new warnings against target Hb levels above 12 g/dL (HCT of 36%) for “all patients.” The FDA has also issued specific warnings against off-label use in cancer patients whose anemia is not directly linked to chemotherapy. The FDA has consistently reminded physicians that the main endpoint in studies for on-label indications has been avoidance of or reduction in transfusions.

All ESAs covered in this LCD per FDA indications must be administered per FDA-approved label guidance. Please see the FDA drug label for the FDA-approved indications and dosages. This can be accessed at: https://labels.fda.gov/.

Centers for Medicare and Medicaid Services (CMS) has issued a National Coverage Determination (NCD) 110.21 for the use of ESAs in cancer and related neoplastic conditions.

Please refer to CMS Internet-Only Manual Publication 100-03, Medicare National Coverage Determinations (NCD) Manual, Chapter 1, Part 2, §110.21 for nationally covered indications related to ESA treatment for anemia secondary to myelosuppressive anticancer chemotherapy in solid tumors, multiple myeloma, lymphoma, and lymphocytic leukemia. This national determination includes dosing information which is not superseded by this LCD. Nationally non-covered indications for which ESA treatment is not reasonable and necessary for beneficiaries with certain clinical conditions is also detailed. The Decision Memo for ESAs for non-renal disease indications (CAG-00383N) provides insight related to the evidence analysis use in promulgating the NCD.

This LCD does not supersede but does incorporate information from the NCD and covers some additional limited non-cancer related indications per the discretion afforded to Medicare Administrative Contractors (MACs). For specificity as to billing and coding advice that will support the reasonable and necessary nature of ESA administration for various conditions, it is critical that the related billing and coding article to this LCD be reviewed.

This A/B MAC does recognize the widely variable use of ESAs for a broad spectrum of conditions. The strength and quantity of evidence supporting such uses is also widely variable in terms of quality and volume. As such, this LCD has specified only limited coverage outside of FDA-approved indications. Denials of claims related to this limited coverage may be appealed on a case-by-case basis.

Covered Indications for ESAs:

  • Treatment of significant anemia in patients with non-myeloid malignancies where anemia is specifically due to concomitantly administered chemotherapy;
  • Treatment of symptomatic anemia related to end-stage renal disease (ESRD) and stages IIIb, IV and V chronic kidney disease (CKD);
  • Treatment of anemia induced by AZT (Zidovudine) used in HIV/AIDS therapy;
  • Treatment of selected patients with anemia related to low prognostic risk myelodysplastic syndrome (MDS) and some myeloproliferative neoplasms;
  • Peri-surgical adjuvant therapy for purposes of allogenic RBC transfusion reduction

The following causes of anemia should be considered, documented, and corrected before starting or continuing ESA therapy for any of the above covered indications:

  • Iron deficiency;
  • Underlying infection, inflammatory or malignant processes;
  • Underlying hematological disease;
  • Hemolysis;
  • Vitamin deficiencies (e.g., folic acid or B12);
  • Blood loss-overt or occult;
  • Aluminum intoxication;
  • Osteitis fibrosis cystica; or
  • Pure RBC aplasia

ESA treatment is not reasonable and necessary for beneficiaries with certain clinical conditions, either because of a deleterious effect of the ESA on their underlying disease or because the underlying disease increases their risk of adverse effects related to ESA use. These conditions include:

  • Any anemia in cancer or cancer treatment patients due to folate deficiency, B-12 deficiency, iron deficiency, hemolysis, bleeding, or bone marrow fibrosis;
  • The anemia associated with the treatment of acute and chronic myelogenous leukemias (chronic myeloid leukemia (CML), acute myeloid leukemia (AML)), or erythroid cancers;
  • The anemia of cancer not related to cancer treatment;
  • Any anemia associated only with radiotherapy;
  • Prophylactic use to prevent chemotherapy-induced anemia;
  • Prophylactic use to reduce tumor hypoxia;
  • Patients with EPO-type resistance due to neutralizing antibodies; and
  • Anemia due to cancer treatment if patients have uncontrolled hypertension.

Non-ESRD ESA services are not considered reasonable and necessary within the context of other medical conditions for which resolution would be reasonably expected prior to starting or continuing ESA administration. Such conditions would include, but not be limited to iron/vitamin B12/folate deficiencies, G6PD deficiency, pyridoxine deficiency, various forms of hemolysis, hereditary spherocytosis, and pure red cell aplasias. The presence of any of these conditions would reduce the therapeutic impact and effectiveness of the ESA. Additionally, the presence of unspecified anemia suggests appropriate evaluation, to determine the nature of the treated anemia, has not been completed.

There are very rare patients whose cardiac, pulmonary or other medical conditions warrant the use of ESAs to maintain a Hb/HCT higher than the FDA target levels discussed in this LCD. Documentation to support this practice must be available upon request. (This instruction does not apply to ESA therapy for anemia related to cancer chemotherapy, which follows the rules mandated by the NCD 110.21.)

During therapy with an ESA, many patients will require supplemental iron. For these patients, stores of iron should be regularly monitored. Reference the related billing and coding article for further detail.

For patients receiving chemotherapy for non-myeloid malignancies, the goal of therapy is to avoid transfusions. ESA therapy will be reimbursed only when the Hb is less than 10 g/dL or the HCT is less than 30%.

For all other indications, the goal of therapy is to maintain a stable Hb and HCT, with target ranges of 10-12 g/dL and 30-36% respectively. Doses must be titrated according to the patient’s response. ESA therapy need not be stopped completely simply due to the achievement of the target Hb and/or HCT. However, judicious, appropriately timed dose adjustments are expected to prevent inappropriate increases in Hb and HCT levels.

The likelihood of anemia associated with EPO deficiency increases as renal failure progresses and the diseased kidneys are unable to produce sufficient amounts of EPO. The anemia of CKD should not be confused with the anemia of chronic disease. In the latter, inflammatory cytokines suppress the endogenous production of EPO and erythropoiesis directly. Measurable levels of circulating cytokines may be found in stable dialysis patients, but, in the absence of inflammation, do not adversely affect the action of ESAs. In patients with impaired renal function and a normochromic, normocytic anemia, it is rare for the serum EPO level to be elevated. Therefore, measurement of EPO levels in such patients is not likely to guide clinical decision making or ESA therapy.

ESAs may be administered by intravenous or subcutaneous routes. An intravenous route is generally recommended for an ESRD indication. Please see the related billing and coding article for details related to required modifier use on claims for ESA administration reporting.

Coverage Criteria: (Review the related billing and coding article for further detail regarding documentation)

  1. For ESRD patients on dialysis
    • Diagnosis of ESRD
    • Anemia of ESRD with a Hb < 10 gm/dL or a HCT < 30% at initiation of therapy
    • Most recent creatinine within the past month prior to initiation or next dosing of ESA
    • Use of an ESA that is FDA-approved for this indication
  2. For CKD patients NOT on dialysis
    • Anemia of CKD with a Hb < 10 g/dL or a HCT < 30% at initiation of therapy
    • Most recent creatinine within the past month prior to initiation or next dosing of ESA
    • Glomerular filtration rate (GFR) less than 45 mL/min/1.73 m2
    • Use of an ESA that is FDA-approved for this indication
  3. For patients with non-myeloid malignancies with anemia due to chemotherapy

    This LCD does not replace, modify or supersede existing Medicare applicable NCDs.

    • Hb level immediately prior to initiation or maintenance of ESA treatment is < 10 g/dL (or HCT is < 30%)
    • Use of an ESA that is FDA-approved for this indication
    • The starting dose for ESA treatment is the recommended FDA label starting dose.
    • Maintenance of ESA therapy is the starting dose if the Hb level remains below 10 g/dL (or HCT is < 30%) 4 weeks after initiation of therapy and the rise in Hb is > 1g/dL (HCT > 3%).
    • For patients whose Hb rises < 1g/dL (HCT rise < 3%) compared to pretreatment baseline over 4 weeks of treatment and whose Hb remains < 10 g/dL afterwards (or HCT < 30%), the recommended FDA label starting dose may be increased once by 25%. Continued use of the drug is not reasonable and necessary if the Hb rises < 1g/dL (HCT rise < 3%) compared to pretreatment baseline by 8 weeks of treatment.
    • Continued administration of the drug is not reasonable and necessary if there is a rapid rise in Hb > 1 g/dl (HCT >3%) over 2 weeks of treatment unless the Hb or HCT remains below or subsequently falls to < 10 g/dL or 30% respectively. Continuation and reinstitution of ESA therapy must include a dose reduction of 25% from the previously administered dose.
    • ESA treatment duration for each course of chemotherapy includes the 8 weeks following the final dose of myelosuppressive chemotherapy in a chemotherapy regimen.
  4. For patients with anemia related to AZT treatment for HIV/AIDS
    • Anemia with Hb < 10 g/dL or a HCT < 30% at initiation of therapy
    • Use of an ESA that is FDA-approved for this indication
    • An AZT dose < 4200 mg/week
    • An endogenous baseline pre-transfusion serum EPO (sEPO) level < 500 mU/mL
  5. For Peri-surgical adjuvant therapy to reduce allogenic transfusion:
    • Undergoing planned elective major hip or knee surgery
    • Pre-surgical anemia with Hb between 10 and 13 g/dL at least 3 weeks prior to surgery
    • Use of an ESA that is FDA-approved for this indication
    • Not a candidate for autologous blood transfusion
    • Expectation for peri-operative blood loss of 2 units or more
    • Previous evaluation to ensure that the existing anemia is likely due to chronic disease rather than another reversible condition 
      • In addition to the FDA-labeled indications above, ESAs are covered for the following off-label indications. At this time, there is no clearly established dosing regimen for off-label indications.

  6.  For patients with symptomatic anemia related to very low, low or low score intermediate risk MDS 
    • Revised International Prognostic Scoring System (IPSS-R) score correlating to very low, low or a low score intermediate risk or IPSS score of low or intermediate-1 risk or WPSS of very low, low or intermediate risk*
    • Pretreatment EPO levels of 500 or less
    • Documented anemia-related symptoms such as fatigue, pallor, infection, bleeding or bruising or transfusion dependence
    • Documentation of a reasonable expectancy of longer survival with a reduced need for transfusion support
    • Diagnosis of MDS confirmed by bone marrow aspiration and/or biopsy report
    • Anemia with Hb < 10 g/dL or a HCT of < 30% at initiation of therapy. If after 2 months of treatment, there is no significant increase in Hb/HCT and/or a significant decrease in transfusion requirements, EPO analog therapy should be stopped. 

Limitations Specified by CMS and/or This A/B MAC

See the related billing and coding article for further detail regarding necessary coding information.

* This A/B MAC will monitor new developing prognostic stratification systems, especially those anticipated to be based on mutational analysis and will adjust the billing and coding article as needed in this regard.

 

Summary of Evidence

ESA Efficacy

The United States (U.S.) Normal HCT Trial by Besarab, et al.1 was the first of a series of randomized controlled trials (RCTs) which cast serious doubt on the assumption that full anemia correction should be achieved in the majority of dialysis patients. A cohort of 1233 prevalent CKD5 HD patients with symptomatic heart failure or ischemic heart disease were allocated to either partial treatment of anemia or full anemia correction, using epoetin-alfa. The eventually achieved HCT values were 31% and 40%, respectively. In the normal HCT group treated with epoetin there were 183 deaths and 19 myocardial infarcts, producing 202 primary events, compared to 164 events (150 deaths, 14 myocardial infarcts) in the group in which anemia was partially corrected with epoetin. The risk ratio for the primary endpoint was 1.3 (95% confidence interval [CI] 0.9–1.9) which did not satisfy the pre-specified criterion for statistical significance (even though the nominal p value was 0.03) after adjusting for interim analyses. The trial was stopped early in a situation where the primary hypothesis was unlikely to be proven and the intervention being tested caused harm: 39% had vascular access clotting in the intervention arm and 29% in the control arm (P=0.001).

The double-blind Canada-Europe trial by Parfrey, et al.2 of 596 incident CKD5 HD patients without symptomatic heart disease (18% with diabetic nephropathy) examined the question whether full anemia correction by epoetin-alfa in the group randomized to a Hb target of 13.5–14.5 g/dl, as compared to partial treatment of anemia in the group randomized to a Hb target of 9.5–11.5 g/dl, had a beneficial effect on left ventricular volume and mass index. The eventually achieved Hb values were 13.1 and 10.8 g/dl, respectively. There was no difference in left ventricular volume index or mass index between the 2 groups during this 96-week study. Of note, patients in the full anemia correction group had a significantly higher stroke incidence (secondary endpoint) than patients in the partial treatment correction group. However, the absolute numbers of patients with stroke were very small. As one might expect, the high Hb group received significantly fewer transfusions than the low Hb group, but extent of the benefit was modest: although 9% in the high Hb arm received at least 1 transfusion compared to 19% in the low Hb arm (P=0.004) during the 96-week study, the transfusions per patient per year was 0.3 in the high Hb arm and 0.7 in the low Hb arm (P<0.0001).

The U.S. CHOIR study by Singh, et al.3 similarly aimed to show superiority of full anemia correction by ESA administration in terms of cardiovascular events and death, as compared to partial treatment of anemia, in patients with CKD not yet on dialysis. In this trial, 1432 CKD 3–4 patients (49% with diabetes) were randomized to Hb targets of 13.5 g/dl and 11.3 g/dl using epoetin alfa. Withdrawal rate was high: 17% due to renal replacement therapy and 21% for other reasons. The study was prematurely stopped after an interim analysis with a median study duration of 16 months. The achieved Hb values were 12.6 and 11.3 g/dl, respectively. At this time point, 125 patients in the complete anemia correction group but only 97 patients in the standard correction group had reached the primary combined cardiovascular endpoint (P=0.03). No differences in quality of life (QoL) were observed comparing the 2 groups; although, again, this finding must be interpreted cautiously because the study was open label.

Finally, the international trial of darbepoetin-alfa in type 2 diabetes and CKD (TREAT) by Pfeffer, et al.4 examined cardiovascular and kidney outcomes in 4038 CKD 3–4 patients. Of note, this is by far the largest ESA trial, and has the best research design, as it was placebo-controlled and double-blinded. Patients received either darbepoetin-alfa to achieve a Hb target of 13.0 g/dl or placebo with rescue darbepoetin-alfa when the Hb concentration was < 9.0 g/dl. The achieved Hb values were 12.5 and 10.6 g/dl, respectively. The median follow-up duration of the study was 29 months. There were no differences in the 2 primary endpoints, which were the composite outcomes of death or a cardiovascular event (first primary endpoint) and death or ESRD (second primary endpoint). The hazard ratio for death/composite cardiovascular event was 1.05 (95% CI 0.94–1.17), and for death or ESRD it was 1.06 (95% CI 0.96–1.19). However, there was a substantial increased risk of stroke (HR 1.92; 95% CI 1.38–2.68), although the absolute risk of stroke overall was modest: 5.0% of the high Hb group had a stroke compared to 2.6% in the placebo group (P< 0.001). The relative increase in risk of stroke was similar in patients with and without a past history of stroke. As a result, the absolute risk of stroke was substantial in the 11% of subjects with a prior history of stroke; 12% in the darbepoetin group compared to 4% in the placebo group. Venous thrombo-embolic events occurred significantly more frequently in the high Hb arm (2.0%) compared to the placebo arm (1.1%, P=0.02). A signal that normalization of Hb with darbepoetin may be harmful in patients with a history of malignancy was reported following a post-hoc analysis: 14/188 (7.4%) of those with a history of malignancy at baseline died from cancer in the darbepoetin arm compared to 1/160 (0.6%) (P=0.002) in the placebo arm. A statistically significant improvement in Functional Assessment of Cancer Therapy-Fatigue (FACT-fatigue) scores was reported at week 26 favoring the darbepoetin group, but the clinical significance of this was modest as 55% of the high Hb group had a clinically important improvement in fatigue score compared to 50% of the placebo group. Transfusions were prescribed relatively frequently and more often in the placebo arm (25%) compared to the high Hb arm (15%). The harm:benefit trade-off in TREAT was 1 stroke for 5 transfusions prevented by the high Hb target. In a large subset of the TREAT patients, QoL was assessed using FACT-fatigue, 36-Item Short Form Survey Instrument (SF-36), and EQ-5D through 97 weeks. Compared to placebo, darbepoetin conferred a consistent, but small improvement over 97 weeks in fatigue and overall QoL, but none in energy and physical function.

Lab Analysis in Renal Patients Treated with ESAs

The Kidney Disease: Improving Global Outcomes (KDIGO) 2012 Anemia Guidelines-Chapter 3 (Use of ESAs and other agents to treat anemia in CKD) 5 regarding frequency of monitoring was reviewed. Recommendations 3.12.1-3.12.3 noted Hb measurement at least monthly during initiation of ESAs. For maintenance therapy in CKD non-dialysis patients, measurement is recommended at least every 3 months and for CKD 5 dialysis patients, at least monthly. Other key recommendations for ESA treatment included the following:

  • Address all correctable causes of anemia (including iron deficiency and inflammatory states) prior to initiation of ESA therapy. (Not Graded)
  • In initiating and maintaining ESA therapy, potential benefits of reducing blood transfusions and anemia-related symptoms must be balanced against the risks of harm in individual patients (e.g., stroke, vascular access loss, hypertension). (1B)
  • Use of ESA therapy with great caution, if at all, in CKD patients with active malignancy, in particular when cure is the anticipated outcome (1B), a history of stroke (1B), or a history of malignancy (2C).

Fishbane and Spinowitz in the Core Curriculum 2018: Update on Anemia in ESRD and Earlier Stages of CKD6 extensively review both the considerable frequency and complexity of iron metabolism in CKD. Certainly before starting ESA treatment, iron status should be optimized. After initiating ESA treatment, the recommendation is made for weekly Hb testing until Hb stability and goals are achieved. A reasonable goal of an increase of 1 g/dL within the first month of treatment is noted. As Hb concentration increases, a large amount of iron is transferred from storage tissues to the developing red cells and iron deficiency is frequently induced. Since this may limit the effectiveness of ESA treatment, these authors of this nephrology fellowship core education curriculum, recommend monthly iron status testing during the initial ESA phase.

Hsu, et al. (2001)7 set out to better delineate relationships between level of renal function and magnitude of reduction in HCT. This study was a cross-sectional one examining 12,055 (8495 women, 3560 men) adult patients with outpatient medical records at Brigham and Women’s Hospital. Racial categorization for this population was ~ 45% white, 25% black, and 30% other. Age averaged 50. Instances of apparent AKI were excluded. Since the anemia of CKD is characterized by normocytic/normochromic RBCs, the analysis was also limited to subjects with RBC parameters falling within normal reference ranges. In this study, renal function was assessed in 2 ways: the Cockcroft-Gault equation for estimated creatinine clearance (CrCl) and Modification of Diet in Renal Disease (MDRD) formula for GFR estimation normalized to body surface area. Results showed that HCT decreased progressively below estimated CrCl 60 mL/min in men and 40 mL/min in women. Compared with male subjects who had a CrCl > 80 mL/min, men with CrCl 60 mL/min had a 1% lower mean HCT, in those with a CrCl 50-40 mL/min the mean HCT was 2.4% lower, in those with a CrCl 40-30 mL/min the mean HCT was 3.7% lower, in those with a CrCl 30-20 mL/min the mean HCT was 3.5% lower, and in those with a CrCl < 20 mL/min the mean HCT reduction was 10% lower. For women, the HCT significantly decreased starting at a CrCl of less than 40 mL/min. With a CrCl of 40-30 mL/min the mean HCT was 1.7% lower, with a CrCl of 30-20 mL/min the mean HCT was 2.9% lower, and at a CrCl < 20 mL/min the mean HCT was 6.3% lower. All these values had a P value < 0.05. These results were similar with a slightly diminished gender difference when indexed to body size. Men with GFRs of 50-40 mL/min/1.73m2, 40-30 mL/min/1.73m2, 30-20 mL/min/1.73m2, and < 20 mL/min/1.73m2 had mean HCTs that were lower by 2.0, 4.4, 5.3, and 9.4% respectively (P < 0.05). Corresponding reductions in women with GFR 50-40 mL/min/1.73m2, 40-30 mL/min/1.73m2, 30-20 mL/min/1.73m2, and < 20 mL/min/1.73m2 were 0.6, 1.6, 3.8, and 5.3% (P < 0.05). For the entirety of this study, the results were similar if Hb was used instead of HCT. Further stratified analysis did not reveal effect modification by race or age. The analyses were limited by smaller numbers of patients in the lowest categories of CrCl and GFR.

Hsu, et al.8 published a separate study the following year which set out to study and quantify the relationship between reduced renal function and Hb level, to assess the iron status of CKD patients, and to estimate the burden of anemia related to CKD. The nationally representative Third National Health and Nutrition Examination Survey (NHANES III) (1988-1994) data of 15,971 patients (8506 women and 7465 men) was utilized with the measurements of creatinine, Hb, and iron stores analyzed. Older people, Mexican Americans and African Americans were oversampled. A statistically significant decrease in Hb was seen in men at CrCl < 70 mL/min and in women starting at < 50 mL/min. At any given level, men had a larger decrease in Hb than women. It was clear that a substantial number of patients with CKD did not have sufficient iron stores to support erythropoiesis. In those patients with CrCl 20-30 mL/min, 46% of women and 19% of men had transferrin saturation levels < 20%. 47% of women and 44% of men had serum ferritin levels < 100 ng/mL. With specific regard to Hb and renal function, the authors divided the subjects into 8 categories of renal function by their Cockcroft-Gault calculated CrCl: > 80 mL/min, >70 to < 80 mL/min, > 60 to < 70 mL/min, 50 to < 60 mL/min, 40 to < 50 mL/min, 30 to < 40 mL/min, 20 to < 30 mL/min, and < 20 mL/min. Hb was examined as the dependent variable in a general linear model. Three different thresholds were used to define anemia: Hb < 10, < 11, and < 12 g/dL. The likelihood of anemia at different levels of renal function was then linearly modeled. The following CrCl characteristics were noted:

The following table notes the predicted change in mean Hb level by renal function*

CrCl Women Men
Change in Hemoglobin (g/dl) Value Change in Hemoglobin (g/dl) P Value
CrCl > 80 ml/min Reference   Reference  
80 > CrCl > 70 ml/min -0.0 (-0.1, 0.1) 0.68 -0.1 (-0.2, 0.0) 0.16
70 > CrCl > 60 ml/min -0.1 (-0.2, 0.0) 0.08 -0.2 (-0.3, -0.0) 0.02
60 > CrCl > 50 ml/min -0.1 (-0.2, 0.1) 0.36 -0.3 (-0.5, -0.1) 0.007
50 > CrCl > 40 ml/min -0.2 (-0.3, -0.0) 0.01 -0.4 (-0.6, -0.2) 0.0005
40 > CrCl > 30 ml/min -0.4 (-0.6, -0.2) <0.0001 -0.8 (-1.1, -0.6) <0.0001
30 > CrCl > 20 ml/min -1.0 (-1.2, -0.7) <0.0001 -1.4 (-2.1, -0.6) 0.0005
CrCl < 20 ml/min -2.3 (-2.8, -1.9) <0.0001 -2.7 (-3.8, -1.6) <0.0001

*Adjusted for age and race/ethnicity; values in parentheses are 95% confidence intervals for parameter estimates. CrCl, creatinine clearance.

 

And in Table 4 of the study:

Table 4. Predicted likelihood (%) of anemia by renal function in select demographic subgroups (multivariate analysis)

Demographic Subgroup Women Men
Likelihood (5) of Hemoglobin Level: Likelihood (5) of Hemoglobin Level:
<10 g/dl <11 g/dl <12 g/dl <10 g/dl <11 g/dl <12 g/dl
Non-Hispanic white
age 31-40 CrCl > 80 ml/min 2 4 11 <1 <1 1
50 > CrCl > 40 ml/min 3 6 15 <1 1 2
30 > CrCl > 20 ml/min 10 18 34 2 3 6
age 61-70 CrCl > 80 ml/min 1 2 7 1 1 3
50 > CrCl > 40 ml/min 1 3 10 1 2 4
30 > CrCl > 20 ml/min 6 12 25 4 7 12
Non-Hispanic black
age 31-40 CrCl > 80 ml/min 8 15 30 1 2 5
50 > CrCl > 40 ml/min 12 21 38 2 4 7
30 > CrCl > 20 ml/min 29 42 63 7 10 17
age 61-70 CrCl > 80 ml/min 5 10 22 3 5 9
50 > CrCl > 40 ml/min 7 14 28 5 8 13
30 > CrCl > 20 ml/min 20 32 52 13 18 28
Mexican-American
age 31-40 CrCl > 80 ml/min 3 6 16 <1 <1 1
50 > CrCl > 40 ml/min 5 10 21 <1 1 2
30 > CrCl > 20 ml/min 15 24 43 2 3 5
age 61-70 CrCl > 80 ml/min 2 4 10 1 1 2
50 > CrCl > 40 ml/min 3 6 14 1 2 4
30 > CrCl > 20 ml/min 9 17 32 4 6 10

* CrCl, creatinine clearance.

 

Compared with subjects with a CrCl > 80mL/min, the decrease in Hb for subjects with CrCl of 20-30 mL/min was 1 g/dL in women and 1.4 g/dL in men.

NKF-K/DOQI Clinical Practice Guidelines5 recommend an evaluation of anemia among patients with CKD when the Hb is < 11g/dL among premenopausal women and < 12 g/dL among adult men and postmenopausal women. ESA treatment is recommended, whether dialysis-dependent or not, to achieve a target Hb of 11-12 g/dL.

El-Achkar, et al.10 evaluated Kidney Early Evaluation Program (KEEP) data in a cross-sectional, community-based study to determine the prevalence of anemia by level of kidney function and by diabetes status. While the latter connection was the focus, the stratified data in this study specific to estimated GFR correlation with Hb levels was reviewed for purposes of this LCD. GFR was calculated using serum creatinine values and categorized as > 90, 60-89, 30-59, and < 30 mL/min/1.73m2. Anemia was defined as Hb < 12 g/dL in men and women older than 50, and < 11 g/dL in women < 50. 5380 subjects were included with mean age of 52.5, race of 43% black/36% white/11% Hispanic/11% other, and 33% male/67% female. 814 subjects were staged CKD 3. Our attention was drawn to the data detail table demonstrating the distribution of anemia prevalence by K/DOQI categories and 10 mL/min/1.73m2 increments of estimated GFR. 40.4% of diabetics in GFR category 31-40 had anemia, 23.5% of diabetics in GFR 41-50 had anemia, and 16.2% in GFR 51-60 had anemia. For non-diabetic subjects, 20.8% of GFR 31-40 were anemic, 10.1% of GFR 41-50 were anemic and 4.8% of GFR 51-60 were anemic. The table detailing mean Hb for the 10 mL/min/1.73m2 increments of estimated GRF showed the following results for CKD stage 3:

EGFR Hb for Men w/ DM Hb for Men w/o DM Hb for Women w/ DM Hb for Women w/o DM
51-60 13.9 (P=0.006) 14.7 13.1 13.5
41-50 13.3 (P<0.001) 14.3 12.9 13.2
31-40 12.2 (P<0.001) 14.2 12.4 13.0

In a much older, but still relevant study, Radtke, et al.11 studied 117 patients not in need of dialysis. Five subgroups were created for CrCl between 2-9, 10-19, 20-29, and 30-39 and 40-90 mL/min/1.73m2. With CrCl plotted against the corresponding HCT, renal anemia was clearly manifest in patients with a CrCl below 40 mL/min/1.73m2. In the range between 2 and 40 mL/min/1.73m2, CrCl and HCT were highly significantly correlated (P < 0.0001) whereas there was no significant correlation existing within the range between 41 and 90 mL/min/1.73m2.

Myelodysplastic Syndrome (MDS) and ESA Use

ESAs including EPO, recombinant human EPO and darbepoetin have been evaluated in the treatment of symptomatic anemia in patients with MDS. Studies in mostly lower risk MDS patients have demonstrated response rates of 40% and 60% in initial trials. Clinical trial results have noted overall response rates to darbepoetin similar to or possibly higher than epoetin. These improved response rates may be related to dosage used (150-300 mcg SQ per week) or to the fact that better-risk patients were enrolled. Predictive features for a better ESA response rate have included low basal serum erythropoietin (sEpo) levels, low percentages of marrow blasts, and fewer prior blood transfusions.

A study from the French myelodysplasia group12 analyzed ESA outcomes (epoetin or darbepoetin), with or without growth colony stimulating factor (G-CSF), in 403 MDS patients with anemia. Based on the International Work Group (IWG) 2000 criteria, the hematologic response rate was 62% with a median duration of 20 months. Based on the IWG 2006 criteria, the hematologic response rate was 50% for a median duration of 24 months. IPSS low- or intermediate-1 (int-1) risk was associated with significantly higher response rates and longer response durations. In comparing outcomes for low vs. int-1 risk subsets with anemia between those treated (N=284) and a historical cohort of untreated patients (N=225), multivariate analysis showed a significant association between treatment with ESAs and survival outcomes.

A phase II study13 evaluated darbepoetin (given Q 2 weeks for 12 weeks), with or without G-CSF (added at 12 weeks in non-responders). The lower risk IPSS group with anemia and sEpo levels < 500 mU/mL had hematologic response rates of 48% at 12 weeks and 56% at 24 weeks. This study also showed QoL improvements among patients with responding disease.

The data is more limited regarding the effectiveness of ESAs in the treatment of anemia in lower-risk patients with del(5q). Retrospective studies from the French MDS group reported hematologic response rates between 46% and 64% with median response duration of 11 months (mean duration, 13-14 months) in patients with del(5q) treated with ESAs, with or without G-CSF.12 Duration of response in these patients was significantly decreased compared to patients without del(5q) (mean duration, 25-27 months). Multivariate analysis showed del(5q) was a significant predictor of a shorter response duration with treatment per the French group.

The 2021 NCCN guidelines14 call attention to the fact that the FDA alerts issued in 2007 and 2008 related to observed increased mortality and possible tumor promotion and thromboembolic events did not involve analysis of MDS patients receiving ESAs. The guidelines note that ESAs have been used safely in large numbers of adult MDS patients and have an important role in improvement of anemia related symptoms related to MDS, often with a decrease in blood transfusion requirements. Studies assessing the long-term use of EPO with or without G-CSF in MDS patients have not shown a negative impact on survival or AML evolution when compared to randomized controls or historical controls.

Jadersten, et al.15 reported improved survival in low-risk MDS patients with low transfusion need after treatment with ESAs. The French group study showed improved survival and decreased AML progression of IPSS low or int-1 risk patients after EPO treatment, with or without G-CSF, compared to the historical control International Myelodysplasia Risk Analysis Workshop (IMRAW) database patients. This is further evidence of lack of a negative impact of ESAs in the treatment of MDS. The NCCN panel has thus recommended the use of ESAs in the management of symptomatic anemia in MDS patients, with a target Hb range of 10-12 g/dL. CMS, in constructing the NCD on the use of ESAs in non-renal disease specifically narrowed the scope of the NCD to cancer and related neoplastic conditions which excluded MDS as it is defined as a premalignant condition rather than an oncologic disease. Thus, local Medicare contractors may continue to make reasonable and necessary determinations on the use of ESAs in MDS and MPN (myeloproliferative neoplasms).

The NCCN Guidelines Panel14 recommends lenalidomide be considered for patients with symptomatically anemic non-del(5q) MDS with anemia that did not respond to initial therapy.

The NCCN Guidelines Panel14 recommends therapeutic options for lower-risk patients based on stratification centered on del(5q) status. Patients with del(5q) abnormalities alone or with 1 other cytogenic abnormality, except those involving chromosome 7, and symptomatic anemia should receive lenalidomide. An alternative option to lenalidomide in these patients includes an initial trial of ESAs only when sEpo levels are < 500 mU/mL. In patients without del(5q) status, alone or with 1 other cytogenetic abnormality and with symptomatic anemia, ESA treatment is recommended if the sEpo level is less than or equal to 500 mU/mL, with or without G-CSF. Patients with normal cytogenetics, < 15% ring sideroblasts and sEpo levels < 500 mU/mL may respond to ESAs if relatively high doses are administered. The EPO dose required is 40,000 to 60,000 SC units 1-2 times per week. Darbepoetin should be given SQ at a dose of 150-300 mcg every other week. Erythroid responses generally are evident within 6-8 weeks.

The NCCN Guidelines14 are clear that iron repletion must be verified before instituting ESA therapy.

Prognostic Stratification in MDS

While there are several risk-based stratifications available, the NCCN Guidelines13 preference for IPSS-R has been noted. The IPSS-R system was derived from analysis of a large dataset from multiple international institutions. It refines the original IPSS by providing more detail for cytogenetic subgroups, detailing subgroups within the “marrow blasts < 5%” group and providing more precision regarding cutoffs for the cytopenias. Other parameters including age, performance status, serum ferritin, LDH and beta-2 microglobulin provide additional prognostic information for survival outcomes. The predictive value of the IPSS-R has been validated in several independent studies based on registration data reviewed by the NCCN workgroup. Other prognostic stratification systems may be used (e.g., IPSS or WPSS) but ESA treatment is generally most effective for patients with very low, low or low intermediate prognostic risk.

The NCCN Guidelines13 reference a multiregional study of MDS patient registry data from Italy (N=646) Messa, et al. (2012) in which the predictive power of the IPSS-R was found to be greater than the IPSS, WPSS and refined WPSS for OS, AML evolution and progression free survival. The Guidelines also reference the Spanish Group of MDS (2012) who performed a retrospective analysis of data from lower-risk MDS (IPSS low or int-1) patients (N=2410) in Spain in which the application of the IPSS-R system appeared to provide prognostic refinement within the IPSS int-1 group with a large proportion of patients (511 of 1096) identified as having poor prognosis (median survival of 21-30 months). IPSS-R performed better in this regard than the refined WPSS (47% vs. 17%). Per the Guidelines, data from the MDS Clinical Research Consortium also demonstrated improved prognostic value of the IPSS-R in 370 AML transforming MDS patients as compared to the IPPS, the global MD Anderson risk model or the transforming MDS MD Anderson models.

Analysis of Evidence (Rationale for Determination)

The evidence reviewed and the stringent nature of FDA oversight over decades certainly support the FDA label indications16,17,18 and warnings in place for the various ESAs. The core of this policy regarding ESRD and CKD, presurgical, and AZT treatment applications rests upon treatment in concordance with FDA labeling.

NCD 110.21 is the source of detail for the use of ESAs in cancer and related neoplastic conditions.

The literature covering the use of ESAs for anemia of chronic disease is mixed. Most reported studies are small, and positive effects must be balanced with newer data that shows the attendant potential thrombogenic and hypertensive risk. Additionally, some patients given ESAs with anemia of cancer have shorter survival times. Until further publications show clear benefit, ESAs for anemia of chronic diseases will not be covered.

For the delineation of appropriate coverage criteria related to use of ESAs in ESRD and CKD (specifically stages 3b or worse), the referenced 2002 Hsu study was viewed as compelling. The limitations of this study were realized and included use of calculated CrCl rather than actual measurements of CrCl or GFR. It was understood that this was a cross-sectional study and thus not viewing Hb changes over time. The oversampling of some patients at higher risk, such as the elderly and minorities, was viewed as potentially very representative of a Medicare demographic. A representative number of subjects in the stage II and III range of CKD was felt to be present and provided solid data for This A/B MAC evidence review. More than any other study we identified, this one separated renal function into much smaller delineated groups for purposes of gauging a reasonable cut-off value to support a reasonable and necessary standard for ESA use in CKD that truly captures a population who will most likely benefit while also being exposed to the least potential harm. Our review of the stratified CrCl categorizations and the positive correlating relationship of worsening renal function to worsening anemia led to this A/B MAC's conclusion that coverage confined to CKD stages 3b and worse would amply and logically allow for coverage of reasonable and necessary circumstances.

The El-Achkar, et al. study, while specifically designed to analyze the influence of diabetes impact on anemia in CKD patients, also identified a significant correlation between HCT and CrCl occurring below 40 mL/min/1.73m2 in Radtke, et al. and below eGFRs of 50 mL/min/1.73m2 in men and women in the Hsu, et al. studies.

For the purposes of this LCD, this A/B MAC has considerable confidence in the ESA coverage requirements with regard to stage of CKD and Hb/HCT parameters. Iron storage monitoring information within specialty society guidelines (such as NKF-K/DOQI) is confined to a monthly frequency during the initiation phase of ESAs. This LCD establishes monitoring of iron stores during ESA maintenance therapy at least every 3 months as a thoughtful reasonable standard.

Again, detailed coding and billing instructions can be found in the companion local coverage article for this determination.

For the single off-label use of ESAs in the treatment of symptomatic anemia related to very low, low and low scored intermediate risk MDS specified in this LCD, per CMS-issued discretion provided to the MAC, the NCCN Guidelines were used as the foundation to this LCD.

Since more accurate risk stratification by the IPSS-R compared to the IPSS and WPSS has been demonstrated, the IPSS-R categorization is preferred as per NCCN guidelines. Specifically, analysis of patients in the International Working Group for the Prognosis of MDS database, which generated the IPSS-R, has indicated that optimal prognostic separation of lower vs. higher risk patients was obtained based on a dichotomization centered on 3.5 scoring points of the IPSS-R raw score. However, as noted above other prognostic systems may be utilized including those that are developed based on mutational analysis. The evidence basis for use of ESAs in MDS currently best supports coverage when risk is very low, low or low intermediate.

Proposed Process Information

Synopsis of Changes
Changes Fields Changed
N/A
Associated Information
Sources of Information
Bibliography
Open Meetings
Meeting Date Meeting States Meeting Information
N/A
Contractor Advisory Committee (CAC) Meetings
Meeting Date Meeting States Meeting Information
N/A
MAC Meeting Information URLs
N/A
Proposed LCD Posting Date
Comment Period Start Date
Comment Period End Date
Reason for Proposed LCD
Requestor Information
This request was MAC initiated.
Requestor Name Requestor Letter
N/A
Contact for Comments on Proposed LCD

Coding Information

Bill Type Codes

Code Description
N/A

Revenue Codes

Code Description
N/A

CPT/HCPCS Codes

Group 1

Group 1 Paragraph

N/A

Group 1 Codes

N/A

N/A

ICD-10-CM Codes that Support Medical Necessity

Group 1

Group 1 Paragraph:

N/A

Group 1 Codes:

N/A

N/A

ICD-10-CM Codes that DO NOT Support Medical Necessity

Group 1

Group 1 Paragraph:

N/A

Group 1 Codes:

N/A

N/A

Additional ICD-10 Information

General Information

Associated Information

For patients receiving any ESA:

  • The patient's medical record must contain documentation that fully supports the medical necessity for services included within this LCD. This documentation includes, but is not limited to, relevant medical history, physical examination, and results of pertinent diagnostic tests or procedures. The indication for ESA administration must be evident within the medical record. The companion local coverage article to this LCD should be specifically reviewed.
  • Medical record documentation must be legible, maintained in the patient’s medical record, meet the criteria contained in this LCD, and meet billing/coding instruction provided in the accompanying local coverage article.
  • Medical records such as physician’s (or non-physician practitioner's) order must be made available upon request. The medical record should reference current patient weight in kilograms, the ESA units/kg dose, and medical justification for any ESA doses that differ from FDA label instructions. The record must also include pretreatment Hb/HCT results and iron storage laboratory results. In the case of low iron stores, the medical record should include documentation regarding the treatment that was given in terms of repletion. Iron stores, as evidenced by a transferrin saturation of at least 20% and a ferritin level at least > 100 ng/mL must be resulted in order to continue with ESA treatment.
  • Evidence-based medical literature describes a significant increase in ESA administration risk associated with hematocrit levels > 36%. Prompt and judicious dose adjustments are anticipated in response to reaching the target hemoglobin or hematocrit and those efforts must be clearly documented within the medical record. Any delayed reductions or reductions of less than 25% must be justified within the medical record.
  • For Dialysis Patients:
    Documentation must include dialysis schedule and the hemoglobin or hematocrit results immediately prior to the billing period. For ESRD patients on home dialysis, the following additional information must be maintained in the medical record and available to the MAC upon request: a care plan, evidence of home monitoring (including a record of the ESA supplied to the patient and a record of dose administered), patient instructions and patient selection protocol.
  • For Non-dialysis Patients:
    For CKD patients: documentation must include stable baseline eGFR with the accompanying accurate, most specific stage of disease per ICD-10-CM.
  • For patients with symptomatic anemia related to their MDS: the medical record must include the diagnostic bone marrow biopsy report, the date of initiation of ESA therapy, and documentation of the response to ESA administration via changes in Hb/HCT measurements and/or transfusion requirements.

Again, please refer to the related Billing and Coding: Erythropoiesis Stimulating Agents (A58982) article for more granular detail on billing/coding/documentation related to the reasonable and necessary provisions outlined in this LCD.

Sources of Information
N/A
Bibliography
  1. Besarab A, Bolton WK, Browne JK, et al. The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. N Engl J Med. 1998;339(9):584-590.
  2. Parfrey PS, Foley RN, Wittreich BH, Sullivan DJ, Zagari MJ, Frei D. Double-blind comparison of full and partial anemia correction in incident hemodialysis patients without symptomatic heart disease. J Am Soc Nephrol. 2005;16(7):2180-2189.
  3. Singh AK, Szczech L, Tang KL, et al. Correction of anemia with epoetin alfa in chronic kidney disease. N Engl J Med. 2006;355(20):2085-2098.
  4. Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med. 2009;361(21):2019-2032.
  5. KDOQI. National Kidney Foundation clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney disease in adults. Am J Kidney Dis. 2006; 47(5 Suppl 3):S11-S145
  6. Fishbane S, Spinowitz B. Update on anemia in ESRD and earlier stages of CKD: Core curriculum 2018. Am J Kidney Dis. 2018;71(3):423-435.
  7. Hsu CY, Bates DW, Kuperman GJ, Curhan GC. Relationship between hematocrit and renal function in men and women. Kidney Int. 2001;59(2):725-731.
  8. Hsu CY, McCulloch CE, Curhan GC. Epidemiology of anemia associated with chronic renal insufficiency among adults in the United States: Results from the Third National Health and Nutrition Examination Survey. J Am Soc Nephrol. 2002;13(2):504-510.
  9. Retacrit™ (epoetin alfa-epbx). Lake Forest, IL: Hospira, Inc.; 2018. Accessed 12/12/23.
  10. El-Achkar TM, Ohmit SE, McCullough PA, et al. Higher prevalence of anemia with diabetes mellitus in moderate kidney insufficiency: The Kidney Early Evaluation Program. Kidney Int. 2005;67(4):1483-1488.
  11. Radtke HW, Claussner A, Erbes PM, Scheuermann EH, Schoeppe W, Koch KM. Serum erythropoietin concentration in chronic renal failure: Relationship to degree of anemia and excretory renal function. Blood. 1979;54(4):877-884.
  12. Park S, Grabar S, Kelaidi C, et al. Predictive factors of response and survival in myelodysplastic syndrome treated with erythropoietin and G-CSF: The GFM experience. Blood. 2008;111(2):574-582.
  13. Kelaidi C, Beyne-Rauzy O, Braun T, et al. High response rate and improved exercise capacity and quality of life with a new regimen of darbepoetin alfa with or without filgrastim in lower-risk myelodysplastic syndromes: A phase II study by the GFM. Ann Hematol. 2013;92(5):621-631.
  14. National Comprehensive Cancer Network® (NCCN). Guidelines for Treatment of Cancer by Type: Myelodysplastic Syndromes, Version 3.2021-January 15, 2021. Accessed 12/12/23.
  15. Jadersten M, Malcovati L, Dybedal I, et al. Erythropoietin and granulocyte-colony stimulating factor treatment associated with improved survival in myelodysplastic syndrome. J Clin Oncol. 2008;26(21):3607-3613.
  16. Aranesp® (darbepoetin alfa). Thousand Oaks, CA: Amgen® Inc.; 2016. Accessed 12/12/23.
  17. Epogen® (Epoetin alfa). Thousand Oaks, CA: Amgen® Inc.; 2007. Accessed 12/12/23.

Revision History Information

Revision History Date Revision History Number Revision History Explanation Reasons for Change
01/18/2024 R1

Under CMS National Coverage Policy section headings were updated for regulations. The following regulations were removed and placed in the related Billing and Coding article: Title XVIII of the Social Security Act, §1842(u) and §1881(b)(11)(B)(i); CMS Internet-Only Manual, Pub. 100-02, Chapter 6, §10.2 and §30, and Chapter 11, §30.1; CMS Internet-Only Manual, Pub. 100-04, Chapter 4, §10, §20, §50.1 and §200.2, Chapter 6, §20, §20.2.1 and §20.2.1.1, Chapter 7, §100, Chapter 8, §10.5, §40, §50.2, §50.3, §60.2, §60.2.1.1, §60.2.1.2 and §60.4, Chapter 17, §10, §20.5.8, §40.1 and §80.5, and Chapter 30. The following regulations were removed due to no longer being available: CMS Internet-Only Manual, Pub. 100-04, Chapter 8, §50.1.5, §50.2.5, §60.4.2.1, §60.4.2.2, §60.4.3.1 and §60.4.3.2. The following regulations were removed due to already being included in the related Billing and Coding article: CMS Internet-Only Manual, Pub. 100-02, Chapter 11, §20.3; CMS Internet-Only Manual, Pub. 100-04, Chapter 8, §60.4.2, and Chapter 17, §80.8, §80.9, §80.10, §80.11 and §80.12. Under Bibliography reference #9 was deleted as this is a duplicate of reference #5 and remaining references were renumbered accordingly. Changes were made to citations to reflect AMA citation guidelines. Formatting, punctuation and typographical errors were corrected and acronyms were inserted and defined where appropriate throughout the LCD.

  • Provider Education/Guidance
N/A

Associated Documents

Attachments
N/A
Related National Coverage Documents
N/A
Public Versions
Updated On Effective Dates Status
01/12/2024 01/18/2024 - N/A Currently in Effect You are here
06/03/2022 07/24/2022 - 01/17/2024 Superseded View

Keywords

  • Erythropoiesis Stimulating Agents
  • ESAs

Read the LCD Disclaimer