Coverage Policy Manual
Policy #: 2012019
Category: Laboratory
Initiated: May 2012
Last Review: February 2024
  Genetic Test: FLT, NPM1, and CEBPA Variants in Cytogenetically Normal Acute Myeloid Leukemia

Description:
Treatment of acute myeloid leukemia (AML) is based on risk stratification, mainly patient age and tumor cytogenetics. In patients with cytogenetically normal AML, the identification of mutations in several genes, including FLT3, NPM1, and CEBPA, has been proposed to allow for further segregation in the management of this heterogeneous disease.
 
Background
 
Acute Myeloid Leukemia
AML is a group of diverse hematologic malignancies characterized by the clonal expansion of myeloid blasts in the bone marrow, blood and/or other tissues. It is the most common type of leukemia in adults and is generally associated with a poor prognosis. The American Cancer Society has estimated there will be 20,380 new cases of AML and 11,310 deaths from AML in the United States in 2023 (ACS, 2023).
 
Diagnosis and Prognosis of AML
The most recent World Health Organization (WHO) classification (2022) reflects the increasing number of acute leukemias that can be categorized based on underlying cytogenetic abnormalities (i.e., at the level of the chromosome including chromosomal translocations or deletions) or molecular genetic abnormalities (i.e., at the level of the function of individual genes, including gene variants) and those distinguished by differentiation without defining genetic abnormalities. These cytogenetic and molecular changes form distinct clinicopathologic-genetic entities with diagnostic, prognostic, and therapeutic implications (Dohner, 2010). Conventional cytogenetic analysis (karyotyping) is considered to be a mandatory component in the diagnostic evaluation of a patient with suspected acute leukemia because the cytogenetic profile of the tumor is considered to be the most powerful predictor of prognosis in AML and is used to guide the current risk-adapted treatment strategies.
 
Molecular variants have been analyzed to subdivide AML with normal cytogenetics into prognostic subsets. In AML, 3 of the most frequent molecular changes with prognostic impact are variants of CEBPA, encoding a transcription factor, variants of the FLT3 gene, encoding a receptor of tyrosine kinase involved in hematopoiesis and a variant of the NPM1 gene, encoding a shuttle protein within the nucleolus. “AML with NPM1 mutation” and “AML with CEBPA mutation” were included as categories in the 2022 WHO classification of acute leukemias. AML with FLT3 variants is not considered a distinct entity in the 2022 or prior 2016 classifications (Khoury, 2022; Arber, 2016). The 2008 WHO classification recommended determining the presence of FLT3 variants because of the prognostic significance (Dohner, 2010).
 
Treatment
AML has a highly heterogeneous clinical course, and treatment generally depends on the different risk-stratification categories (Liersch, 2014). Depending on the risk-stratification category, treatment modalities may include intensive remission induction chemotherapy, hypomethylating agents, enrollment in clinical trials with innovative compounds, palliative cytotoxic treatment or supportive care only. For patients who achieve a complete remission (CR) after induction treatment, possible postremission treatment options include intensive consolidation therapy, maintenance therapy or autologous or allogeneic hematopoietic stem-cell transplantation (HSCT) (Liersch, 2014).
 
Measurable (Minimal) Residual Disease Monitoring
Relapse in AML is believed to be due to residual clonal cells that remain following "complete response” after induction therapy but are below the limits of detection using conventional morphologic assessment (Ehinger, 2019). Residual clonal cells that can be detected in the bone marrow or blood are referred to as measurable residual disease (MRD), also known as minimal residual disease. MRD assessment is typically performed by multiparameter flow cytometry or polymerase chain reaction with primers for common variants. It is proposed that finding MRD at different time points in the course of the disease (e.g., after initial induction, prior to allogenic transplantation) may be able to identify patients at a higher risk for relapse. In those with a high risk of relapse during the first remission, stem cell transplantation may be a more appropriate treatment approach. Studies in both children and adults with AML have demonstrated the correlation between MRD and risk for relapse. The role of MRD monitoring in AML is evolving and important limitations remain. Some patients may have relapse despite having no MRD, while others do not relapse despite being MRD positive. Standards have recently been introduced for identifying certain individual markers for MRD assessment, and threshold values delineating MRD positivity and negativity have recently been defined for multiparameter flow cytometry and some variants detected by polymerase chain reaction or other methods (Heuser, 2021).
 
FLT3 Variants
FMS-like tyrosine kinase (FLT3) plays a critical role in normal hematopoiesis and cellular growth in hematopoietic stem and progenitor cells. Variants in FLT3 are among the most frequently encountered in AML (Levis, 2013). FLT3 variants are divided into 2 categories: (1) internal tandem duplications (FLT3-ITD) variants, which occur in or near the juxtamembrane domain of the receptor, and (2) point mutations resulting in single amino acid substitutions within the activation loop of the tyrosine kinase domain (FLT3-TKD).
 
FLT3-ITD variants are much more common than FLT3-TKD variants, occurring in 30% of newly diagnosed adult cases of AML, versus FLT3-TKD variants, occurring in about 10% of patients (NCCN, 2022). FLT3-ITD are a well-documented adverse prognostic marker, particularly in patients younger than 60 years of age and with normal or intermediate risk cytogenetics and are associated with an increased risk of relapse and inferior overall survival (OS) (Levis, 2013; Whitman, 2010; Patel, 2012). Patients with FLT3-ITD mutations have a worse prognosis when treated with conventional chemotherapy, compared with patients with wild type (i.e., nonmutated) FLT3. Although remission can be achieved in patients with FLT3-ITD variations using conventional induction chemotherapy at a frequency similar to other AML patients, the remission durations are shorter and relapse rates are higher. The median time to relapse in patients with a FLT3-ITD mutation is 6 to 7 months compared with 9 to 11 months in patients with other AML subtypes (Levis, 2013).
 
Because of the high risk of relapse, hematopoietic stem-cell transplantation (HSCT) as consolidation therapy of the first remission for an FLT3-ITD AML patient is often considered. However, this treatment must be weighed against the treatment-related mortality associated with a transplant (Levis, 2013).
 
The clinical significance of an FLT3 variant varies by the nature of the variant and the context in which it occurs. Longer FLT3-ITD variants have been associated with worse overall survival (Polak, 2022).
 
For FLT3-ITD variants, the allelic ratio refers to the number of ITD-mutated alleles compared with the number of WT (nonmutated) alleles. This ratio is influenced by the number of malignant versus benign cells in the sample tested and by the percentage of cells with 0, 1 or 2 mutated alleles. In most cases, the variants detected at diagnosis is also present at relapse. However, in some cases, as FLT3-ITD positive AML evolves from diagnosis to relapse, the variant present at diagnosis may be absent (or undetectable) at relapse. This is most commonly seen where the mutant allele burden is low (5%-15%) at diagnosis (Levis, 2013). The assays for detecting FLT3-ITD was previously considered to be unsuitable for use as a marker of minimal residual disease (Levis, 2013). Higher mutant to WT allelic ratios have been associated with worse outcomes (Levis, 2013).
 
The prognostic impact of FLT3-TKD variants is less certain and conflicting. Some studies have suggested a negative impact of tyrosine kinase domain variants on event-free survival and overall survival, while other studies have found no prognostic value, or potentially a benefit if a NPM1 mutation is also present (Daver, 2019; Bazarbachi, 2020; NCCN, 2020).
 
NPM1 Variants
A common molecular aberration in AML is a variant of NPM1, which is found in 28% to 35% of AML cases and is more common in cytogenetically normal AML (CN-AML) (NCCN, 2021). Up to 50% of AML with mutated NPM1 also carry a FLT3-ITD. Mutated NPM1 confers an independent favorable prognosis for patients with CN-AML and either the presence or absence of a FLT3-ITD variant. Retrospective studies of banked clinical samples suggest that a NPM1 variant may mitigate the negative prognostic effect of the FLT3-ITD, but possibly only if the FLT3-ITD to WT allelic ratio is low (Levis, 2013). The prognostic impact in patients with an abnormal karyotype is unclear (Liersch, 2014).
 
CEBPA Variants
CEBPA (CCAAT/enhancer-binding protein) is a transcription factor gene that plays a role in cell cycle regulation and cell differentiation. Variants to CEBPA are found in approximately 7% to 11% of AML patients (Martelli, 2013; Ohgami, 2015, NCCN, 2020). CEBPA variants can be either biallelic (double variants) or monoallelic. Monoallelic variants are prognostically similar to CEBPA WT variant and do not confer a favorable prognosis in cytogenetically normal AML; with the exception of mutations in the basic leucine zipper region, double variants of CEBPA and variants with single mutations in the basic leucine zipper region have shown a better prognosis with higher rates of complete remission and overall survival after standard induction chemotherapy (Cagnetta, 2014; Li, 2015; Tarlock, 2021; Taube, 2022).
 
Regulatory Status
Clinical laboratories may develop and validate tests in-house and market them as a laboratory service; such tests must meet the general regulatory standards of the Clinical Laboratory Improvement Act. Several Laboratories offer these tests including Quest Diagnostics, Medical Genetic Laboratories of Baylor College, Geneva Labs of Wisconsin, LabPMM and ARUP Laboratories, and they are available under the auspices of the Clinical Laboratory Improvement Amendments. Laboratories that offer laboratory-developed tests must be licensed under the Clinical Laboratory Improvement Amendments for high-complexity testing. To date, the U.S. Food and Drug Administration has chosen not to require any regulatory review of this test.
 
The FDA has granted approval for midostaurin (Rydapt, Novartis Pharmaceuticals), gilteritinib (Xospata, Astellas Pharma US), and quizartinib (Vanflyta, Daiichi Sakyo) for the treatment of acute myeloid leukemia with a FLT3 mutation as detected by an FDA-approved test. A list of cleared or approved companion diagnostic devices can be found at: https://www.fda.gov/medical-devices/in-vitro-diagnostics/list-cleared-or-approved-companion-diagnostic-devices-in-vitro-and-imaging-tools.
 
 
Coding
There is specific CPT coding for the following testing:
 
81218 - CEBPA (CCAAT/enhancer binding protein [C/EBP], alpha) (e.g., acute myeloid leukemia), gene analysis, full gene sequence
 
81245 – FLT3 (fms-related tyrosine kinase 3) (e.g., acute myeloid leukemia), gene analysis; internal tandem duplication (ITD) variants (i.e., exons 14, 15)
 
81246 - FLT3 (fms-related tyrosine kinase 3) (e.g., acute myeloid leukemia), gene analysis; tyrosine kinase domain (TKD) variants (e.g., D835, I836)
 
81310 – NPM1 (nucleophosmin) (e.g., acute myeloid leukemia) gene analysis, exon 12 variants
 
0023U- Oncology (acute myelogenous leukemia), DNA, genotyping of internal tandem duplication, p.D835, p.I836, using mononuclear cells, reported as detection or non-detection of FLT3 mutation and indication for or against the use of midostaurin (new code effective 10/01/17).
 
81347 - SF3B1 (splicing factor [3b] subunit B1) (e.g., myelodysplastic syndrome/acute myeloid leukemia) gene analysis, common variants (e.g., A672T, E622D, L833F, R625C, R625L) (new code effective 1/1/2021)
 
81348 - SRSF2 (serine and arginine rich splicing factor 2) (e.g., myelodysplastic syndrome, acute myeloid leukemia) gene analysis, common variants (e.g., P95H, P95L) (new code effective 1/1/2021)

Policy/
Coverage:
Effective January 2021
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Genetic testing for FLT3 internal tandem duplication (FLT3/ITD), NPM1 and CEBPA variants meets member benefit certificate primary coverage criteria of effectiveness in cytogenetically normal AML to guide management decisions in patients who would receive treatment other than low-dose chemotherapy or best supportive care (e.g, high-dose chemotherapy)
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Genetic testing for FLT3 internal tandem duplication (FLT3/ITD), NPM1, and CEBPA variants does not meet member benefit certificate primary coverage criteria of effectiveness in all other situations.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3 internal tandem duplication (FLT3/ITD), NPM1 and CEBPA variants is considered investigational in all other situations.
 
Genetic testing for FLT3 tyrosine kinase domain (FLT3/TKD) variants does not meet member benefit certificate primary coverage criteria of effectiveness.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3 tyrosine kinase domain (FLT3/TKD) variants is considered investigational.
 
Genetic testing for FLT3, NPM1 and CEBPA variants to detect minimal residual disease does not meet member benefit certificate primary coverage criteria of effectiveness.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3, NPM1 and CEBPA variants to detect minimal residual disease is considered investigational.
 
Genetic testing for SF3B1 common variants (e.g. A672T, E622D, L833F, R625C, R625L) (eg, myelodysplastic syndrome, acute myeloid leukemia) does not meet member benefit certificate primary coverage criteria of effectiveness.
 
For members with contracts without primary coverage criteria, genetic testing SF3B1 common variants (e.g. A672T, E622D, L833F, R625C, R625L) (eg, myelodysplastic syndrome, acute myeloid leukemia) is considered investigational.
 
Genetic testing for SRSF2  common variants (eg, P95H, P95L) (eg, myelodysplastic syndrome, acute myeloid leukemia) does not meet member benefit certificate primary coverage criteria of effectiveness.
 
For members with contracts without primary coverage criteria, genetic testing SRSF2  common variants (eg, P95H, P95L) (eg, myelodysplastic syndrome, acute myeloid leukemia) is considered investigational.
 
 
Effective Prior to January 2021
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Genetic testing for FLT3 internal tandem duplication (FLT3/ITD), NPM1 and CEBPA variants meets member benefit certificate primary coverage criteria of effectiveness in cytogenetically normal AML to guide management decisions in patients who would receive treatment other than low-dose chemotherapy or best supportive care (e.g, high-dose chemotherapy)
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Genetic testing for FLT3 internal tandem duplication (FLT3/ITD), NPM1, and CEBPA variants does not meet member benefit certificate primary coverage criteria of effectiveness in all other situations.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3 internal tandem duplication (FLT3/ITD), NPM1 and CEBPA variants is considered investigational in all other situations.
 
Genetic testing for FLT3 tyrosine kinase domain (FLT3/TKD) variants does not meet member benefit certificate primary coverage criteria of effectiveness.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3 tyrosine kinase domain (FLT3/TKD) variants is considered investigational.
 
Genetic testing for FLT3, NPM1 and CEBPA variants to detect minimal residual disease does not meet member benefit certificate primary coverage criteria of effectiveness.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3, NPM1 and CEBPA variants to detect minimal residual disease is considered investigational.
 
Effective August 2014 – February 2018
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Genetic testing for FLT3 internal tandem duplication (FLT3/ITD) and NPM1 mutations meets member benefit certificate primary coverage criteria of effectiveness in cytogenetically normal AML to guide management decisions in patients who would receive treatment other than low-dose chemotherapy or best supportive care (e.g, high-dose chemotherapy)
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Genetic testing for FLT3 internal tandem duplication (FLT3/ITD) and NPM1 does not meet member benefit certificate primary coverage criteria of effectiveness in all other situations.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3 internal tandem duplication (FLT3/ITD) and NPM1 mutations is considered investigational in all other situations.
 
Genetic testing for FLT3 tyrosine kinase domain (FLT3/TKD) mutations does not meet member benefit certificate primary coverage criteria of effectiveness.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3 tyrosine kinase domain (FLT3/TKD) mutations is considered investigational.
 
Genetic testing for FLT3 or NPM1 mutations to detect minimal residual disease does not meet member benefit certificate primary coverage criteria of effectiveness.
 
For members with contracts without primary coverage criteria, genetic testing for FLT3 or NPM1 mutations to detect minimal residual disease is considered investigational.
 
 
 
Effective prior to August 2014
Testing for FLT3 and NPM1 mutations to predict response to high-dose chemotherapy in patients diagnosed with acute myelogenous leukemia meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes.
 

Rationale:
Analytic validity (technical accuracy of the test in detecting a mutation that is present or in excluding a mutation that is absent)
 
No published data on the analytic validity of FLT3 or NPM1 mutation testing is identified.
 
Clinical validity (diagnostic performance of the test [sensitivity, specificity, positive and negative predictive values] in detecting clinical disease)
 
Published data on the clinical validity of FLT3 testing is lacking, however, a review article highlights that a major limitation of most polymerase chain reaction (PCR) assays for FLT3/ITD mutations is lack of sensitivity, compared with PCR assays for other acute myeloid leukemia (AML)‒associated genetic alterations (Levis, 2013). The sensitivity of the PCR assays is a function of the amount of sample DNA and the number of PCR cycles. However, for the FLT3/ITD assay, increasing the number of cycles does not increase the sensitivity because the PCR primers used to amplify the mutant allele also amplify the wild-type (WT) allele, and the shorter WT allele has a competitive advantage over the mutant allele, because it takes more time to complete a PCR cycle for the longer-length mutant allele. The longer the mutation (insertion), the greater the PCR bias (Levis, 2013).
 
This bias can be minimized using fewer PCR cycles, but this could affect the sensitivity if there is a low burden of leukemia cells in the sample (Levis, 2013).
 
Published data on the clinical validity of testing for NPM1 mutations is not identified.
 
Clinical utility (how the results of the diagnostic test will be used to change management of the patient and whether these changes in management lead to clinically important improvements in health outcomes)
 
The clinical utility of molecular testing for FLT3 and NPM1 mutations is in whether such testing will change patient management and if this change in management will lead to improved patient outcomes.
 
The literature on the use of these markers consists of retrospective analyses, and no prospective studies have been published to date.
 
Most of the literature consists of analyses of FLT3/ITD mutations and survival outcomes with the use of allogeneic hematopoietic stem-cell transplantations (HSCT) in patients depending on the presence of this type of mutation. In general, the data support the use of HSCT in patients with FLT3/ITD mutations, however, not all studies have shown consistent results (Levis, 2013).
 
Gale et al first reported the results of a retrospective analysis of FLT3 status in patients enrolled in 2 trials in the United Kingdom (Gale, 2005). The trials included 1135 adult patients with AML, of whom 141 received autologous HSCT and 170 an allogeneic HSCT in first complete remission (CR), based on donor availability. An FLT3/ITD was detected in 283 of the total study population of 1135. Of the patients who underwent autologous HSCT (n=141), 37 (26%) were FLT3/ITD-positive and among those who received an allogeneic HSCT (n=170), 35 (21%) were FLT3/ITD-positive. The clinical investigators were not aware of FLT3/ITD status and did not direct treatment based on FLT3 mutation status. There was no difference in effect on relapse rate with the use of autologous versus allogeneic HSCT (odds ratio [OR], 2.39; confidence interval [CI], 1.24 to 4.62 for autologous; OR=1.31; CI, 0.56 to 3.06 for allogeneic; p=0.3), nor between patients who did or did not receive a transplant (p=0.4).
 
They performed an additional analysis of the effect of allogeneic HSCT in FLT3/ITD-positive patients, by performing a donor versus no donor analysis of 683 patients in whom FLT3/ITD status was available. No difference in relapse rate was noted in FLT3/ITD-positive versus negative patients (OR=0.70; CI, 0.53 to 0.92 vs OR=0.59; CI, 0.40 to 0.87; respectively; p=.5). The authors concluded that their results suggest that there is no strong evidence that FLT3 status should influence the decision whether to proceed to transplant.
 
In 2012, Brunet et al retrospectively compared outcomes for FLT3/ITD AML patients registered in the European Group for Blood and Marrow Transplantation (EBMT) who underwent a myeloablative allogeneic HSCT in first remission, compared with patients without the mutation (Brunet, 2012).  Of 1467 patients who met inclusion criteria (age 18 years or older, de novo AML, normal cytogenetics at diagnosis, myeloablative allogeneic HSCT performed between 2000 and 2008), 206 (14%) had FLT/ITD data. FLT/ITD was present in 120 patients, absent in 86. At 2 years, the relapse incidence was 30%±5% versus 16%±5% (p=0.006) in FLT3/ITD-positive versus FLT3/ITD-negative patients, and leukemia-free survival (LFS) 58%±5% versus 71%±6% (p=0.04), in FLT3-positive patients versus negative, respectively. Although the presence of FLT3/ITD led to a higher relapse risk and inferior LFS in this study when compared with the FLT3-negative patients, the observed 2-year LFS of 58% and the relapse risk of 30% in the patients with the FLT3/ITD mutation compares favorably with outcomes that have been reported in patients with FLT3/ITD mutations after postremission chemotherapy (ie, that did not undergo transplant), which has been reported to have a median survival of 2.5 months.
 
Bornhäuser et al reported the results of the AML 96 study of the DSIL (German study initiative leukemia) in which 999 patients 60 years of age or younger were prospectively included between 1996 and 2003 and stratified according to cytogenetic risk category (Bornhauser, 2007).  Of patients with intermediate-risk cytogenetics, 555 were available for evaluation of FLT3 mutation status; 175 (31.5%) were FLT3/ITD positive. The rate of remission after 2 cycles of induction chemotherapy including high-dose Ara-C, was not different in patients with and without FLT3/ITD (68% vs 63%). The investigators decided to determine the impact of different consolidation therapies on overall survival (OS) and the probability of relapse with respect to FLT3/ITD mutation status. Patients underwent allogeneic HSCT (n=103), autologous HSCT (n=141) if no donor was available, or conventional consolidation chemotherapy consisting of high-dose Ara-c (n=132) if the patient could not mobilize autologous cells. After a median follow-up of 53 months, OS was not significantly different between FLT3/ITD positive and negative patients having undergone autologous or allogeneic HSCT. In the group that received conventional consolidation chemotherapy, FLT3/ITD-positive patients had an inferior probability of survival (21% vs 46%; hazard ratio [HR], 2.2; 95% CI, 1.4 to 3.5; p=0.001), and the relapse probability was significantly higher in FLT3/ITD-positive versus negative patients (94% vs 59%; HR=4.0; 95% CI, 2.5 to 6.6; p<0.001).
 
Dezern et al reviewed the clinical data from November 2004 to October 2008 of 133 consecutive patients with previously untreated AML (DeZem, 2011). Patients were between the ages of 20 and 59 and received induction and consolidation therapy at Johns Hopkins, and were followed through August 2010. Thirty-one patients (23%) harbored an FLT3/ITD mutation. Induction success was similar between the 2 groups with 20 of 31 (65%) of FLT3/ITD mutation patients and 52/85 (61%) of WT patients. Of the 20 FLT3/ITD patients in complete remission (CR1), 11 (55%) underwent allogeneic HSCT, 9 myeloablative and 2 nonmyeloablative. The FLT3/ITD patients who did not undergo HSCT either did not have a suitable donor or had precluding comorbidities. Seventeen (33%) of the WT patients underwent HSCT in CR1; 14 myeloablative, 1 syngeneic, 1 autologous and 1 nonmyeloablative allogeneic. In the FLT3/ITD nontransplant group, median relapse-free survival was 8.6 months (range, 5.3-43.3 months) versus 54.1 months (range, 6.4-69.9 months) in the FLT3/ITD transplant group (p=0.03). Median OS in the WT, nontransplant group versus the WT, transplant group was 57.3 months (range, 3.9-64.4) versus 60 months, respectively (p=0.02). The authors conclude that their study suggests an advantage of HSCT in patients with FLT3/ITD in early CR1. However, the number of patients transplanted was small
 
Willemze et al conducted a randomized trial in 1942 newly diagnosed patients with AML, age 15 to 60 years to compare remission induction treatment containing either standard or high-dose cytarabine (Willemze, 2014). In both arms, patients who achieved CR received consolidation therapy with either an autologous or allogeneic HSCT. Patients were subclassified as good risk, intermediate risk, bad risk, very bad risk or unknown risk, according to cytogenetics and FLT3/ITD mutation. Testing for FLT3/ITD mutation showed that in the standard dose cytarabine group, 50% were negative, 13% were positive, and 37% were unknown. In the high-dose cytarabine group, 48% were negative, 14% were positive, and 38% were unknown. All patients with a FLT/ITD mutation were categorized as very bad risk. OS at 6 years in the patients categorized as very bad risk was 20% in the standard cytarabine group and 31% in the high-dose group (HR=0.70; 95% CI, 0.47 to 1.04; p=0.02). The authors concluded that patients with very bad risk cytogenetics and/or FLT3/ITD mutation benefitted from high-dose cytarabine induction treatment.
 
Pratcorona et al reported on the outcomes of 303 patients with intermediate-risk cytogenetics AML who were treated with intensive chemotherapy (Pratcorona, 2013). They analyzed the effect of the ratio of FLT3/ITD to FLT3 WT, depending on the presence of an NPM1 mutation. FLT3/ITD mutations were identified in 94 (31%) of patients and NPM1 mutations in 161 (53%) of patients (65 patients harbored both mutations). To further confirm the prognostic value of the FLT3/ITD mutations to WT ratio, the patients were also subdivided into FLT3wt, FLT3-ITD/wt, ratio <0.5 (low ratio) and FLT3-ITD/wt ratio 0.5 (high ratio). The 0.5 cutoff value was chosen based on maximum clinical prognostic impact derived at that threshold as, in this series, this cutoff showed the greatest difference in relapse rate in patients with FLT3/ITD. Among the patients with NPM1 mutations, FLTwt and low ratio groups showed similar OS, relapse risk, and LFS. High ratio patients had a worse outcome. In patients without NPM1 mutations, FLT3/ITD subgroups showed comparable outcomes, with a higher risk of relapse and shortened OS than WT FLT3 patients.
 
Clinical Trials
No ongoing phase 3 trials comparing the outcomes of allogeneic or autologous HSCT depending on FLT3 or NPM1 mutation status are identified.
 
Summary
Acute myeloid leukemia (AML) is a heterogeneous disease and treatment is based on risk stratification, mainly by patient age and tumor cytogenetics (karyotyping), which allow for patients to be divided into good, intermediate, and poor risk categories. The identification of mutations in several genes, including FLT3 and NPM, have been proposed to allow for further segregation of prognostic categories in the cytogenetically normal group.
 
FLT/ITD mutations are known to confer a very poor prognosis, whereas NPM1 mutations have been shown to confer an independently favorable prognosis, and limited data suggest that a coexistent NPM1 mutation may mitigate the negative prognostic effect of an FLT/ITD mutation, if both mutations are present. The prognostic effect of FLT/TKD mutations is uncertain.
 
Data on the analytic and clinical validity of FLT3 and NPM1 mutation testing are lacking. Data on the clinical utility of testing for these mutations is limited to retrospective analyses, and consist predominantly of studies of the effect of the presence of a FLT3/ITD mutation in patients who underwent hematopoietic stem-cell transplant versus those who did not. Although some controversy exists as to the survival benefit in transplanting a patient with an FLT3/ITD mutation, retrospective studies, in general, have suggested a survival benefit in transplanting these poor risk patients, and major professional societies and guidelines recommend testing for these mutations for risk stratification and treatment management decisions, including possible hematopoietic stem-cell transplantation.
 
Therefore, genetic testing for FLT3 internal tandem duplication (FLT3/ITD) and NPM1 mutations may be considered medically necessary in cytogenetically normal AML, whereas genetic testing for FLT3 tyrosine kinase domain (FLT3/TKD) mutations is considered investigational.
 
Practice Guidelines
The 2014 National Comprehensive Cancer Network guidelines for Acute Myeloid Leukemia(NCCN, 2014) (v 2.2014) provide the following recommendations:
 
For the evaluation and initial workup for suspected acute leukemias, bone marrow analysis with cytogenetics (karyotype) with or without fluorescence in situ hybridization (FISH) is necessary to establish the diagnosis of AML; cryopreservation of samples for evaluation of other markers, including FLT3-ITD and NPM1 mutations.
 
Evaluation of several molecular markers (e.g., FLT3, NPM1, CEBPA, and c-KIT) may be important for risk assessment and prognostication, and may also guide treatment decisions.
 
The American Society of Clinical Oncology states that classification of AML increasing relies on genetic analysis and that broad-based mutation profiling of AML will be helpful in defining important prognostic subgroups and may contribute to the selection of patients for enrollment into trials with novel inhibitors.
 
The Alberta Provincial Hematology Tumour Team issued a 2009 guideline on acute myeloid leukemia that includes the recommendation for molecular analysis in cases with normal karyotypes, including FMS-like tyrosine kinase 3 (FLT3).
 
2016 Update
 
A literature search conducted through November 2016 did not reveal any new published literature that would prompt a change in the coverage statement.
 
2017 Update
A literature search conducted using the MEDLINE database through October 2017 did not reveal any new information that would prompt a change in the coverage statement.  
 
2019 Update
A literature search was conducted through May 2019.  There was no new information identified that would prompt a change in the coverage statement.  
 
2020 Update
A literature search was conducted through May 2020.  There was no new information identified that would prompt a change in the coverage statement.  
 
2021 Update
Annual policy review completed with a literature search using the MEDLINE database through May 2021. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Prognosis of patients with FLT3 internal tandem duplication (ITD) wild type variant was compared to patients without FLT3-ITD variants in a systematic review of 10 studies published between 1999 to 2020 (Rinaldi, 2020). 1,513 adult, non-transplant patients with AML participated in the study. Results include: Overall Survival HR=1.91 (95% CI, 1.59 to 2.30) and event-free survival HR=1.64 (95% CI, 1.26 to 2.14).
 
Monitoring for measurable residual disease (MRD) can provide prognostic information on the risk of relapse in patients with NPM1-mutated AML. Results of studies evaluating the use of MRD with this variant is summarized below (Ivey, 2016; Balsat, 2017; Dillon, 2020).
 
Ivey et al conducted a retrospective evaluation of samples obtained from patients who had undergone intensive treatment in the National Cancer Research Institute AML17 trial (April 2009 to May 2012), with a prospective evaluation period (June 2012 to December 2014) to make up a validation cohort (Ivey, 2016). 346 patients with NPM1-mutated AML participated. The outcome for positive MRD status vs negative MRD status in peripheral blood following the second chemotherapy cycle (retrospective cohort) include: Risk of relapse at 3 years: 82% vs 30% (HR=4.80 [95% CI, 2.95 to 7.80]) and OS at 3 years: 24% vs 75% (HR=4.38 [95% CI, 2.57 to 7.47]). The outcome for positive MRD status vs negative MRD status in peripheral blood following the second chemotherapy cycle (validation cohort) include: Risk of relapse at 2 years: 70% vs 31% (p=0.001) and OS at 2 years: 40% vs 87% (p=0.001).
 
Balsat el al conducted a retrospective evaluation of samples obtained from patients who were enrolled in the ALFA-0702 trial (April 2009 to August 2013) (Balsat, 2017). The study included 152 patients with NPM1-mutated AML who achieved complete remission/complete remission with incomplete platelet recovery after induction. Patients with <4-log reduction in NPM1 from baseline vs those with >5-log reduction in NPM1 from baseline had the following outcomes: 3-year cumulative incidence of relapse (CIR): 65.8% vs 20.5% and 3-year OS: 40.8% vs 93.1%.
 
Dillon et al conducted a retrospective evaluation of samples obtained from patients who had undergone intensive treatment in the National Cancer Research Institute AML17 trial (2009 to 2014) (Dillon, 2020). The study included 107 patients with NPM1-mutated AML who underwent an allogenic stem cell transplantation. Results of any detectable MRD vs MRD-negative in pre-transplant samples include: 2-year OS of 45% vs 83% (median OS: 10.5 months vs not reached [HR=3.60; 95% CI, 1.92 to 6.77]). Results of high MRD levels vs low MRD levels (<200 copies in peripheral blood and <1000 copies in bone marrow) vs MRD-negative in pre-transplant samples include 2-year OS of 13% vs 63% vs 83%.For those with low MRD levels, FLT3-ITD variant vs FLT3-ITD wild-type the 2-year OS was 25% vs 77%.
 
Voso et al published a subgroup analysis of the trial evaluating outcomes in patients with the tyrosine kinase domain subtype (Voso, 2020). In this subgroup, 5-year event-free survival was significantly better in the midostaurin group than in the placebo group (45.2% vs 30.1%; hazard ratio [HR], 0.66; 95% confidence interval [CI], 0.45 to 0.99; p=0.044), but 5-year overall survival was similar between the 2 treatment groups (65.9% vs 58.0%; HR, 0.74; 95% CI, 0.44 to 1.23; p=0.244).
 
Perl et al published results from an RCT evaluating patients with relapsed/refractory FLT3-mutated AML who were randomized to gilteritinib (an FLT3 inhibitor) or salvage chemotherapy (Perl, 2019). Patients with the ITD subtype (88.4%), tyrosine kinase domain subtype (8.4%), and both subtypes (1.9%) were included. 60.6% of patients had relapsed disease, with 39.4% had primary refractory disease. Median overall survival and percent of patients achieving complete remission was significantly better with gilteritinib.
 
Cortes et al published results from an RCT evaluating patients with relapsed/refractory FLT3-mutated AML who were randomized to quizartinib (an FLT3 inhibitor) or salvage chemotherapy (Cortes, 2019). Only patients with the FLT3 ITD subtype were included. One third of patients had refractory disease, while the rest had relapsed disease. Overall survival was improved with quizartinib compared to salvage chemotherapy.
 
Bataller et al conducted a retrospective analysis of patients with AML with a NPM1 mutation without unfavorable cytogenetics who were treated based on the CETLAM-12 protocol (Bataller, 2020). MRD was evaluated after each chemotherapy cycle and at 3-month intervals for at least 3 years after CR. Patients with MRD after consolidation or confirmed MRD reappearance after molecular response were defined as molecular failures. After confirmation of molecular failure or an overt morphologic relapse (HemR), allo-HCT was recommended but treatment was at the discretion of the attending physician, which could include salvage chemotherapy. 157 adults with NPM1 mutation AML were included in the CETLAM-12 protocol; 91% achieved CR after 1 or 2 courses of chemotherapy. Outcomes after allo-HCT, patients who developed molecular failure (n=33) vs HemR without prior molecular failure (n=13): 2-year OS of 85.7% vs 42%.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through May 2022. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Current National Comprehensive Cancer Network guidelines for acute myeloid leukemia (AML) (v.3.2021) provide the following recommendations (NCCN, 2021):
 
For the evaluation for acute leukemia, bone marrow core biopsy and aspirate analysis, including immunophenotyping and cytochemistry, are needed to risk stratify patients.
 
“Several gene mutations are associated with specific prognoses in a subset of patients (category 2A) and may guide treatment decisions (category 2B). Presently, c-KIT, FLT3-ITD, FLT3-TKD, NPM1, CEBPA (biallelic), IDH1/IDH2, RUNX1, ASXL1, TP53, BCR-ABL, and PML-RAR alpha are included in this group. All patients should be tested for mutations in these genes, and multiplex gene panels and comprehensive next-generation sequencing (NGS) analysis are recommended for the ongoing management of AML and various phases of treatment. To appropriately stratify therapy options, test results of molecular and cytogenetic analyses of immediately actionable genes or chromosomal abnormalities (eg, CBF, FLT3 [ITD or TKD], NPM1, IDH1, or IDH2) should be expedited."
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through May 2023. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
A retrospective analysis of patients enrolled in 4 clinical trials between 1996 and 2016 was conducted by Tarlock et al (Tarlock, 2021). Participants consisted of 2958 children and young adults with AML (5.4% with CEBPA. mutations in the basic leucine zipper region). CEBPA WT vs. CEBPA biallelic vs. CEBPA single mutation in basic leucine zipper region resulted in a 5-year OS of 61% vs. 81% vs. 89% (p<.001 for WT vs. others; p=.259 for single vs. biallelic mutations) and in a 5-year EFS of 46% vs. 64% vs. 64% (p<.001 for WT vs. others, p=.777 for single vs. biallelic mutations).
 
Taube et all conducted a retrospective analysis of patients enrolled in 4 clinical trials or the Study Alliance Leukemia registry and biorepository (Taube, 2022). Participants included 4708 patients who received intensive chemotherapy followed by risk-stratified consolidation, with the option of HCT for eligible patients (5.1% with CEBPA mutations). Biallelic CEBPA vs. unselected single CEBPA mutation vs. CEBPA-WT had a median OS 103.2 months vs. 21.9 months vs. 19.3 months, p<.001 and median EFS 20.7 months vs. 9.4 months vs. 7.0 months, p<.001. Biallelic CEBPA vs. single mutation in basic leucine zipper region of CEBPA vs. single mutation in transcription activation domain of CEBPA vs. CEBPA-WT has a median OS of 103.2 months vs. 63.3 months vs. 12.7 months vs. 17.9 months and median EFS of 20.7 months vs. 17.1 months vs. 5.7 months vs. 7.0 months. Multivariate analysis indicated CEBPA variants with a single mutation in the basic leucine zipper region were independently associated with prolonged OS (HR, 0.62; 95% CI, 0.42 to 0.92) and EFS (HR, 0.537; 95% CI, 0.37 to 0.77) after controlling for cytogenetic risk group, age, white blood cell count, diagnosis of treatment-related AML, FLT3 mutations, NPM1 mutations, and receipt of allogeneic HCT in first CR.
 
Issa et al conducted a retrospective analysis of patients treated at a single center between 2012 and 2020 (Issa, 2022). Participants included 1722 adults with relapsed or refractory AML (12% with NPM1 mutations). NPM1 WT vs. NPM1 variant resulted in OS of 5.5 months vs. 6.1 months (p=.07) and RFS of 5.6 months vs. 5.5 months (p=.4).
 
Dohner et al conducted a retrospective analysis of patients enrolled in the QUAZAR AML-001 trial (Dohner, 2022). 469 patients age 55 years or older with AML with intermediate- or poor-risk cytogenetics who achieved CR following intensive chemotherapy and were not considered candidates for HCT, and were then randomized to receive maintenance therapy with oral azacitidine or placebo. Patients with NPM1 mutations had OS HR=0.63 (0.41 to 0.98) and RFS HR=0.55 (0.35 to 0.84). Patients with NPM1-WT had median OS 19.6 months vs. 14.6 months (p=.023) and median RFS 7.7 months vs. 4.6 months (p=.003). Patients with FLT3 mutations had median OS 28.2 months vs. 9.7 months (p=.114) and median RFS 23.1 months vs. 4.6 months (p=.032). Patients with FLT3-WT had median OS 24.7 months vs. 15.2 months (p=.013) and median RFS 10.2 months vs. 4.9 months (p=.001). Patients with NPM1 mutations vs. NPM1-WT in the placebo arm had OS HR=0.69 (0.49 to 0.97) and RFS HR=0.65 (0.47 to 0.91). Those in the oral azacitidine arm had OS HR=0.52 (0.36 to 0.75) and RFS HR=0.46 (0.31 to 0.66). Patients with FLT3 mutations vs. FLT3-WT in the placebo arm had OS HR=1.25 (0.83 to 1.89) and those in the oral azacitidine arm had OS HR=0.96 (0.60 to 1.54).
 
Grob et al conducted a retrospective analysis of patients enrolled in 3 clinical trials between 2006 and 2017 (Grob, 2022). Participants included 161 patients with de novo FLT3-ITD AML who achieved CR after induction. Capillary fragment length analysis and confirmation by targeted NGS for FLT3-ITD at diagnosis and targeted NGS for FLT3-ITD MRD assessment in CR; the lower limit of detection of the FLT3-ITD MRD assay ranged from allele frequencies of 0.01% to 0.001%. Patients with FLT3-ITD MRD detected in CR vs. not had a 4-year cumulative incidence of relapse 75% vs. 33% (HR=3.70 [95% CI, 2.31 to 5.94]) and a 4-year OS 31% vs. 57% (HR=2.47 [95% CI, 1.59 to 3.84]). Multivariate analysis indicated FLT3-ITD MRD detected in CR was independently associated with risk of relapse (HR=3.55 [95% CI, 1.92 to 6.56]) and reduced overall survival (HR=2.51 [95% CI, 1.42 to 4.43]) when controlling for age, white blood cell count at diagnosis, NPM1 mutation status at diagnosis, and FLT3-ITD allelic ratio at diagnosis.
 
The European LeukemiaNet international expert panel recommendations for the diagnosis and management of adults with AML were updated in 2017 and again in 2022 (Dohner, 2017; Dohner, 2022). The most recent update reflects the 2022 changes to the World Health Organization classification of AML. The panel recommended that screening for NPM1, CEBPA, and FLT3 variants should be part of the diagnostic workup in patients with cytogenetically normal AML because they define disease categories that can inform treatment decisions.
 
The European LeukemiaNet MRD Working Party is an international expert panel convened with the objective of providing guidelines for technical assessment and clinical use of immunophenotypic and molecular MRD testing in AML; the panel's first consensus recommendations were published in 2018, and updated recommendations were published in 2021 (Schuurhuis, 2018; Heuser, 2021). In the 2021 update, the panel recommended that molecular MRD be assessed by real-time quantitative or digital polymerase chain reaction in patients with NPM1, CBFB-MYH11, or RUNX1-RUNX1T1 mutations, and by MFC in all other patients. NGS-based MRD monitoring is considered by the panel to be "useful to refine prognosis in addition to MFC but, to date, there are insufficient data to recommend NGS-MRD as a stand-alone technique." The panel also defined MRD positivity thresholds according to whether <FC or polymerase chain reaction techniques were used, and provisional MRD positivity thresholds for NGS techniques.
 
2024 Update
Annual policy review completed with a literature search using the MEDLINE database through January 2024. No new literature was identified that would prompt a change in the coverage statement.

CPT/HCPCS:
0023UOncology (acute myelogenous leukemia), DNA, genotyping of internal tandem duplication, p.D835, p.I836, using mononuclear cells, reported as detection or non detection of FLT3 mutation and indication for or against the use of midostaurin
0046UFLT3 (fms related tyrosine kinase 3) (eg, acute myeloid leukemia) internal tandem duplication (ITD) variants, quantitative
0049UNPM1 (nucleophosmin) (eg, acute myeloid leukemia) gene analysis, quantitative
81218CEBPA (CCAAT/enhancer binding protein [C/EBP], alpha) (eg, acute myeloid leukemia), gene analysis, full gene sequence
81245FLT3 (fms related tyrosine kinase 3) (eg, acute myeloid leukemia), gene analysis; internal tandem duplication (ITD) variants (ie, exons 14, 15)
81246FLT3 (fms related tyrosine kinase 3) (eg, acute myeloid leukemia), gene analysis; tyrosine kinase domain (TKD) variants (eg, D835, I836)
81310NPM1 (nucleophosmin) (eg, acute myeloid leukemia) gene analysis, exon 12 variants
81347SF3B1 (splicing factor [3b] subunit B1) (eg, myelodysplastic syndrome/acute myeloid leukemia) gene analysis, common variants (eg, A672T, E622D, L833F, R625C, R625L)
81348SRSF2 (serine and arginine rich splicing factor 2) (eg, myelodysplastic syndrome, acute myeloid leukemia) gene analysis, common variants (eg, P95H, P95L)

References: Acute Myeloid Leukemia. Detailed Guide. American Cancer Society. Available at http://www.cancer.org/Cancer/Leukemia-AcuteMyeloidAML/DetailedGuide/index. Last accessed May 11, 2012.

American Cancer Society (ACS).(2021) What Are the Key Statistics About Acute Myeloid Leukemia? 2021; https://www.cancer.org/cancer/acute-myeloid-leukemia/about/key-statistics.html. Accessed November 17, 2021.

American Cancer Society (ACS).(2022) What Are the Key Statistics About Acute Myeloid Leukemia? 2022; https://www.cancer.org/cancer/acute-myeloid-leukemia/about/key-statistics.html. Accessed November 15, 2022.

American Cancer Society (ACS).(2023) Key Statistics for Acute Myeloid Leukemia (AML). 2023; https://www.cancer.org/cancer/acute-myeloid-leukemia/about/key-statistics.html. Accessed December 4, 2023.

Balsat M, Renneville A, Thomas X, et al.(2017) Postinduction Minimal Residual Disease Predicts Outcome and Benefit From Allogeneic Stem Cell Transplantation in Acute Myeloid Leukemia With NPM1 Mutation: A Study by the Acute Leukemia French Association Group. J Clin Oncol. Jan 10 2017; 35(2): 185-193. PMID 28056203

Bataller A, Onate G, Diaz-Beya M, et al.(2020) Acute myeloid leukemia with NPM1 mutation and favorable European LeukemiaNet category: outcome after preemptive intervention based on measurable residual disease. Br J Haematol. Oct 2020; 191(1): 52-61. PMID 32510599

Bazarbachi A, Bug G, Baron F, et al.(2020) Clinical practice recommendation on hematopoietic stem cell transplantation for acute myeloid leukemia patients with FLT3 -internal tandem duplication: : a position statement from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Haematologica. Jun 2020; 105(6): 1507-1516. PMID 32241850

Bornhauser M, Illmer T, Schaich M et al.(2007) Improved outcome after stem-cell transplantation in FLT3/ITD-positive AML. Blood 2007; 109(5):2264-5; author reply 65.

Brunet S, Labopin M, Esteve J et al.(2012) Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis. J Clin Oncol 2012; 30(7):735-41.

Brunner AM, Li S, Fathi AT, et al.(2016) Haematopoietic cell transplantation with and without sorafenib maintenance for patients with FLT3-ITD acute myeloid leukaemia in first complete remission. Br J Haematol. Nov 2016;175(3):496-504. PMID 27434660

Cagnetta A, Adamia S, Acharya C, et al.(2014) Role of genotype-based approach in the clinical management of adult acute myeloid leukemia with normal cytogenetics. Leuk Res. Jun 2014; 38(6): 649-59. PMID 24726781

Cortes JE, Khaled S, Martinelli G, et al.(2019) Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): a multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. Jul 2019; 20(7): 984-997. PMID 31175001

Daifu T, Kato I, Kozuki K, et al.(2014) The Clinical Utility of Genetic Testing for t(8;16)(p11;p13) in Congenital Acute Myeloid Leukemia. J Pediatr Hematol Oncol. 2014 Jan 1.

Daver N, Schlenk RF, Russell NH, et al.(2019) Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia. Feb 2019; 33(2): 299-312. PMID 30651634

DeZern AE, Sung A, Kim S et al.(2007) Role of allogeneic transplantation for FLT3/ITD acute myeloid leukemia: outcomes from 133 consecutive newly diagnosed patients from a single institution. Biol Blood Marrow Transplant 2011; 17(9):1404-9.

Dillon R, Hills R, Freeman S, et al.(2020) Molecular MRD status and outcome after transplantation in NPM1-mutated AML. Blood. Feb 27 2020; 135(9): 680-688. PMID 31932839

Dohner H, Estey E, Grimwade D, et al.(2017) Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. Jan 26 2017;129(4):424-447. PMID 27895058

Dohner H, Estey EH, Amadori S et al.(2010) Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European Blood 2010; 115(3):453-74.

Döhner H, Wei AH, Appelbaum FR, et al.(2022) Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood. Sep 22 2022; 140(12): 1345-1377. PMID 35797463

Döhner H, Wei AH, Roboz GJ, et al.(2022) Prognostic impact of NPM1 and FLT3 mutations in patients with AML in first remission treated with oral azacitidine. Blood. Oct 13 2022; 140(15): 1674-1685. PMID 35960871

Ehinger M, Pettersson L.(2019) Measurable residual disease testing for personalized treatment of acute myeloid leukemia. APMIS. May 2019; 127(5): 337-351. PMID 30919505

El Ghannam D, Taalab MM, Ghazy HF, et al.(2014) DNMT3A R882 mutations in patients with cytogenetically normal acute myeloid leukemia and myelodysplastic syndrome. Blood Cells Mol Dis. 2014 Jun-Aug:53(1-2):61-6/j.bcmd.2014.01.004 Epub 2014 Feb 8.

Fernandez HF, Sun Z, Yao X, et al.(2009) Anthracycline dose intensification in in acute myeloid leukemia. N Engl J Med 2009;361:1249-59.

Gale RE, Hills R, Kottaridis PD et al.(2005) No evidence that FLT3 status should be considered as an indicator for transplantation in acute myeloid leukemia (AML): an analysis of 1135 patients, excluding acute promyelocytic leukemia, from the UK MRC AML10 and 12 trials. Blood 2005; 106(10):3658-65.

Godley LA,(2012) Profiles in Leukemia. N Engl J Med. 2012 Mar 22;366(12):1079-89. Epu

Grob T, Sanders MA, Vonk CM, et al.(2022) Prognostic Value of FLT3 -Internal Tandem Duplication Residual Disease in Acute Myeloid Leukemia. J Clin Oncol. Oct 31 2022: JCO2200715. PMID 36315929

Heuser M, Freeman SD, Ossenkoppele GJ, et al.(2021) 2021 Update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party. Blood. Dec 30 2021; 138(26): 2753-2767. PMID 34724563

Issa GC, Bidikian A, Venugopal S, et al.(2022) Clinical outcomes associated with NPM1 mutations in patients with relapsed or refractory AML. Blood Adv. Nov 02 2022. PMID 36322818

Ivey A, Hills RK, Simpson MA, et al.(2016) Assessment of Minimal Residual Disease in Standard-Risk AML. N Engl J Med. Feb 04 2016; 374(5): 422-33. PMID 26789727

Khan I, Altman JK, Licht JD.(2012) New strategies in acute myeloid leukemia: redefining prognostic markers to guide therapy. Clin Cancer Res 2012; 18(19):5163-71.

Khasawneh MK, Abdel-Wahab O.(2014) Recent Discoveries in Molecular Chgaracterization of Acute Myeloid Luekemia. Curr Hematol Malig Rep. 2014 Mar 9.

Khoury JD, Solary E, Abla O, et al.(2022) The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia. Jul 2022; 36(7): 1703-1719. PMID 35732831

Knapper S, Russell N, Gilkes A, et al.(2017) A randomized assessment of adding the kinase inhibitor lestaurtinib to first-line chemotherapy for FLT3-mutated AML. Blood. Mar 2 2017;129(9):1143-1154. PMID 27872058

Kuhnl A, Grimwade D.(2012) Molecular markers in acute myeloid leukaemia. Int J Hematol 2012; 96(2):153-63.

Levis M.(2013) FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? Hematology Am Soc Hematol Educ Program 2013; 2013:220-6.

Li HY, Deng DH, Huang Y, et al.(2015) Favorable prognosis of biallelic CEBPA gene mutations in acute myeloid leukemia patients: a meta-analysis. Eur J Haematol. May 2015; 94(5): 439-48. PMID 25227715

Liersch R, Muller-Tidow C, Berdel WE et al.(2014) Prognostic factors for acute myeloid leukaemia in adults--biological significance and clinical use. Br J Haematol 2014; 165(1):17-38.

Löwenberg B, Ossenkoppele GJ, van Putten W, et al.(2009) High-dose daunorubicin in older patients with acute myeloid leukemia. N Engl J Med 2009;361:1235-48.

Martelli MP, Sportoletti P, Tiacci E, et al.(2013) Mutational landscape of AML with normal cytogenetics: biological and clinical implications. Blood Rev. Jan 2013; 27(1): 13-22. PMID 23261068

Meshinchi S, Alonzo TA, Stirewalt DL, et al.(2006) Clinical implications of FLT3 mutations in pediatric AML. Blood. Dec 01 2006;108(12):3654-3661. PMID 16912228

National Comprehensive Cancer Network (NCCN). Acute Myeloid Leukemia V2.2014. 2014. Available online at: http://www.nccn.org/professionals/physician_gls/pdf/aml.pdf. Last accessed May, 2014.

National Comprehensive Cancer Network (NCCN).(2020) NCCN Clinical Practice Guidelines in Oncology: Acute Myeloid Leukemia. Version 2.2021. https://www.nccn.org/professionals/physician_gls/pdf/aml.pdf. Accessed November 22, 2020.

National Comprehensive Cancer Network (NCCN).(2021) NCCN Clinical Practice Guidelines in Oncology: Acute Myeloid Leukemia. Version 3.2021. https://www.nccn.org/professionals/physician_gls/pdf/aml.pdf. Accessed November 17, 2021

National Comprehensive Cancer Network (NCCN).(2022) NCCN Clinical Practice Guidelines in Oncology: Acute Myeloid Leukemia. Version 2.2022. https://www.nccn.org/professionals/physician_gls/pdf/aml.pdf. Accessed November 15, 2022.

Ohgami RS, Ma L, Merker JD, et al.(2015) Next-generation sequencing of acute myeloid leukemia identifies the significance of TP53, U2AF1, ASXL1, and TET2 mutations. Mod Pathol. May 2015; 28(5): 706-14. PMID 25412851

Patel JP, Gönen M, Figueroa ME, et al.(2012) Prognostic relevance of integrated genetic profiling in acute myeloid leukemia N Engl J Med. 2012 Mar 22;366(12):1079-89. Epub 2012 Mar 14.

Peloquin GL, Chen YB, Fathi AT.(2013) The evolving landscape in the therapy of acute myeloid leukemia. Protein Cell 2013; 4(10):735-46.

Perl AE, Martinelli G, Cortes JE, et al.(2019) Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3 -Mutated AML. N Engl J Med. Oct 31 2019; 381(18): 1728-1740. PMID 31665578

Polak TB, Van Rosmalen J, Dirven S, et al.(2022) Association of FLT3-internal tandem duplication length with overall survival in acute myeloid leukemia: a systematic review and meta-analysis. Haematologica. Oct 01 2022; 107(10): 2506-2510. PMID 35796012

Pratcorona M, Brunet S, Nomdedeu J et al.(2013) Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy. Blood 2013; 121(14):2734-8.

Rinaldi I, Louisa M, Wiguna FI, et al.(2020) Prognostic Significance of Fms-Like Tyrosine Kinase 3 Internal Tandem Duplication Mutation in Non-Transplant Adult Patients with Acute Myeloblastic Leukemia: A Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev. Oct 01 2020; 21(10): 2827-2836. PMID 3311253

Schuurhuis GJ, Heuser M, Freeman S, et al.(2018) Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party. Blood. Mar 22 2018; 131(12): 1275-1291. PMID 29330221

Siegel RL, Miller KD, Jemal A.(2020) Cancer statistics, 2020. CA Cancer J Clin. Jan 2020; 70(1): 7-30. PMID 31912902

Stone RM, Mandrekar SJ, Sanford BL, et al.(2017) Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. Aug 3 2017;377(5):454-464. PMID 28644114

Tarlock K, Lamble AJ, Wang YC, et al.(2021) CEBPA-bZip mutations are associated with favorable prognosis in de novo AML: a report from the Children's Oncology Group. Blood. Sep 30 2021; 138(13): 1137-1147. PMID 33951732

Taube F, Georgi JA, Kramer M, et al.(2022) CEBPA mutations in 4708 patients with acute myeloid leukemia: differential impact of bZIP and TAD mutations on outcome. Blood. Jan 06 2022; 139(1): 87-103. PMID 34320176

Voso MT, Larson RA, Jones D, et al.(2020) Midostaurin in patients with acute myeloid leukemia and FLT3-TKD mutations: a subanalysis from the RATIFY trial. Blood Adv. Oct 13 2020; 4(19): 4945-4954. PMID 33049054

Walter MJ, Shen D, Ding L. et al.(2012) Clonal architecture of secondary acute myeloid leukemia. N Engl J Med. 2012 Mar 22;366(12):1090-8. Epub 2012 Mar 14.

Whitman SP, Maharry K, Radmacher MD et al.(2010) FLT3 internal tandem duplication associates with adverse outcome and gene- and microRNA-expression signatures in patients 60 years of age or older with primary cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. Blood 2010; 116(18):3622-6.

Whitman SP, Ruppert AS, Radmacher MD et al.(2008) FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications. Blood 2008; 111(3):1552-9.

Willemze R, Suciu S, Meloni G et al.(2014) High-dose cytarabine in induction treatment improves the outcome of adult patients younger than age 46 years with acute myeloid leukemia: results of the EORTC-GIMEMA AML-12 trial. J Clin Oncol 2014; 32(3):219-28.

Wu X, Feng X, Zhao X, et al.(2016) Prognostic significance of FLT3-ITD in pediatric acute myeloid leukemia: a meta-analysis of cohort studies. Mol Cell Biochem. Sep 2016;420(1-2):121-128. PMID 27435859


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.