section name header

Disease Prologue

Summary

Cancers Staged Using This Staging System

Acute myeloid leukemia

Cancers Not Staged Using This Staging System

These histopathologic types of cancer…Are staged according to the classification for…and can be found in chapter…
Multiple myelomaMultiple myeloma82
Myelodysplastic syndromeNo AJCC staging systemN/A
Myeloproliferative neoplasmNo AJCC staging systemN/A
Chronic lymphocytic leukemiaNon-Hodgkin Lymphomas: Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma79
Acute lymphoblastic leukemia in childrenAcute lymphoblastic leukemia in children83.2
Acute lymphocytic leukemia in adultsAcute lymphocytic leukemia in adults83.3
Chronic myeloid leukemiaChronic myeloid leukemia83.4

Summary of Changes

Leukemia is a new malignancy for the AJCC cancer staging system.

ICD-O-3 Topography Codes

CodeDescription
C42.1Bone marrow

WHO Classification of Tumors

This list includes histology codes and preferred terms from the WHO Classification of Tumors and the International Classification of Diseases for Oncology (ICD-O). Most of the terms in this list represent malignant behavior. For cancer reporting purposes, behavior codes /3 (denoting malignant neoplasms), /2 (denoting in situ neoplasms), and in some cases /1 (denoting neoplasms with uncertain and unknown behavior) may be appended to the 4-digit histology codes to create a complete morphology code.

CodeDescription
9840Acute erythroid leukemia
9861Acute myeloid leukemia, NOS
9865Acute myeloid leukemia with t(6;9)(p23;q34); DEK-NUP214
9866Acute promyelocytic leukemia with t(15;17)(q22;q12); PML-RARA
9867Acute myelomonocytic leukemia
9869Acute myeloid leukemia with inv(3)(q21q26.2) or t(3;3)(q21;q26.2); RPN1-EVI1
9870Acute basophilic leukemia
9871Acute myeloid leukemia with inv(16) (p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11
9872Acute myeloid leukemia with minimal differentiation
9873Acute myeloid leukemia without maturation
9874Acute myeloid leukemia with maturation
9891Acute monoblastic and monocytic leukemia
9895Acute myeloid leukemia with myelodysplasia-related changes
9896Acute myeloid leukemia with t(8;21)(q22;q22); RUNX1-RUNX1T1
9897Acute myeloid leukemia with t(9;11)(p22;q23); MLLT3-MLL
9898Myeloid leukemia associated with Down syndrome
9910Acute megakaryoblastic leukemia
9911Acute myeloid leukemia (megakaryoblastic) with t(1;22)(p13;q13); RBM15-MKL1
9920Therapy-related myeloid neoplasms
9930Myeloid sarcoma
9931Acute panmyelosis with myelofibrosis

International Agency for Research on Cancer, World Health Organization. International Classification of Diseases for Oncology. ICD-O-3-Online.http://codes.iarc.fr/home. Accessed August 16, 2017. Used with permission.

Introduction

Leukemia is a cancer of the hematopoietic system. The disease is subdivided by the tempo of disease (acute vs. chronic) as well as the primary lineage involved (myeloid vs. lymphoid). In general, acute leukemia is characterized by a block in cell differentiation, resulting in a proliferation of immature cells (blasts). In chronic leukemia, the cells at diagnosis appear more normally differentiated but generally are very high in number in the bone marrow and peripheral blood. When leukemia is fatal, it generally is because normal hematopoietic function has been usurped by the leukemia, resulting in thrombocytopenia and neutropenia, leading to death from bleeding or infections.

Further subclassification of each leukemia may depend on specific genetic abnormalities [e.g., the t(9;22) “Philadelphia (Ph) chromosome” in acute lymphoblastic leukemia (ALL)] or cell phenotype type (e.g., B- vs. T-cell ALL). In addition, because the acute leukemias more often involve a larger age spectrum than the chronic leukemias (which are rare in younger people), attempts have been made to subclassify these leukemias into pediatric, adult, and elderly. These age boundaries have changed over the years, and there are no established cutoffs.

Because of the nature of the disease, leukemia does not easily fit into the TNM staging system. Rather, clinically meaningful staging is defined differently for each type of leukemia.

Classification Rules

Clinical Classification

Acute myeloid leukemia (AML) is a hematopoietic malignancy that represents the culmination of genetic and epigenetic alterations in hematopoietic stem/progenitor cells (HSPCs), leading to dysregulation of critical signal transduction pathways and resulting in the expansion of undifferentiated myeloid cells.1 AML may be divided broadly into two general categories, de novo AML and secondary AML. De novo AML develops without obvious antecedent. Secondary AML refers to the evolution of AML after exposure to cytotoxic therapy (t-AML) or to an antecedent hematologic disorder (AHD; e.g., myelodysplastic syndrome [MDS]) associated with distinct karyotypic and molecular alterations, including MLL translocations after exposure to topoisomerase inhibitors or complex karyotypes after treatment with alkylating agents.2 However, it now is apparent that some patients with seemingly de novo AML have genomic alterations reminiscent of those seen in secondary AML. In these patients, the clinical picture typically is similar to that of secondary AML.3

Many somatic karyotypic and molecular alterations have been identified in AML. However, despite the association of many of these alterations with distinct clinical phenotypes, most have no prognostic value, nor do they identify a specific target or a distinct pathway that can be readily exploited for therapeutic intervention.1 The paucity of targets is more notable in childhood AML, particularly because the observed age-associated evolution of molecular alterations reveals a distinct profile for younger children with AML compared with older children and adolescents with AML. Furthermore, the land scape of genetic alterations differs markedly from AML in adults.4,5

AML is diagnosed in very young children and comprises nearly 25% of pediatric leukemias; however, it is far more prevalent in adults and generally is considered a disease of older adults, whose median age at diagnosis is nearly 70 years. The incidence of AML is better appreciated by close evaluation of the most recent Surveillance, Epidemiology, and End Results (SEER) data (http://seer.cancer.gov/statfacts/html/amyl.html). These data demonstrate a low incidence of AML in children and young adults, with a significant increase in older adults.6 However, closer examination demonstrates that the highest incidence of AML in younger patients (<40 years) occurs in infancy, with an incidence of 1.6 cases per 100,000, similar to that in the fourth decade of life. After infancy, there is a declining incidence of approximately 0.12 cases per 100,000 per year in the first decade of life to an incidence of 0.4 per 100,000 by age 10. In the following three decades, there is a steady increase in AML incidence of approximately 0.02 cases per 100,000 per year to a rate of 1.3 cases per 100,000 per year by age 45, nearly equivalent to that seen in infants. A substantial rise in the incidence of AML occurs in the fifth decade, reaching nearly 10 times the observed rate in the previous three decades to an incidence of 6.2 per 100,000 by age 65. After age 65, AML diagnosis again increases substantially, more than 30-fold higher than that seen in younger patients (age 10-40). Seemingly spontaneously arising AML in patients over age 60 to 65 often is characterized by complex cytogenetic changes that are very similar to those seen in patients with MDS or in AML arising after receipt of chemotherapy for other conditions.7 This of course suggests that these “spontaneous” AMLs reflect evolution from an underlying MDS and /or the cumulative effect of lifetime toxin exposure. It has been demonstrated that the small minority (5-10%) of patients who receive chemotherapy for other diseases and subsequently develop AML have specific polymorphisms for genes involved in detoxifying carcinogens.8-10

It is becoming increasingly apparent that aging is accompanied by development of aberrations in genes that are also abnormal in AML, such as DNMT3a and ASXL.11 This observation led to the suggestion that acquisition of AML, like other cancers, is a multistep process that may take years to complete. Indeed, the genetic land scape of AML is complex. An average of 13 coding mutations have been reported in de novo AML, with, on average, five of these occurring in genes that are recurrently mutated, consistent with a pathogenic role in AML.1 Most mutations appear to be stochastically acquired events in normal HSPCs, and these are retained after the HSPC acquires a relatively small number of “driver” mutation(s). However, this small number ignores observations that during AML evolution, multiple new critical abnormalities may be acquired that will then act as active drivers in disease progression.12 These additional mutations may be downstream of the initial (founding) mutation(s), or they may bypass these founding mutations by using a parallel cellular pathway. These processes may be hastened by AML chemotherapy. In this fashion, the founding clone frequently gives rise to a variety of subclones potentially resistant to therapy. These clones may predominate at relapse, making AML, molecularly at least, a “progressive” disease.

Laboratory Features

Unlike cancers of solid organs, AML is widely disseminated at diagnosis; hence, a TNM-type staging system is of little practical value. For many years, classification of AML was almost exclusively morphologic and was dominated by the French-American-British (FAB) system.13 AML was diagnosed only if the marrow or blood contained >30% myeloid blasts, identified primarily by histochemical stains specific to, for example, the granulocytic (peroxidase) or monocytic lineages (esterase). Various FAB subtypes, M0 through M7, were recognized. A problem with the FAB system was that its clinical relevance was limited, with the very important exception of subtype M3, acute promyelocytic leukemia (APL), which unlike other types of AML, is routinely curable with all-trans retinoic acid and arsenic trioxide with or without an anthracycline such as idarubicin. Furthermore, as more was learned about the prognostic significance of cytogenetics and various molecular abnormalities, it became obvious that any classification system needed to incorporate this information. This recognition led to development of the World Health Organization (WHO) system, currently the most widely accepted means of classifying AML.14

Unlike the FAB system, the WHO criterion for a diagnosis of AML is >20% blasts, with the exceptions noted here. This change from >30% reflected observations that after accounting for various prognostic factors, such as cytogenetics or whether AML was de novo or secondary (unfavorable), patients with 21-30% blasts fared identically to those with >30% blasts when treated identically.15 Some experts have contended, however, that any minimum blast criterion (many cooperative groups use 10%) is too simplistic and ignores the complex medical decision making inherent in AML treatment. Rather than relying on histochemical stains to identify blasts of myeloid lineage, cell surface antigens now serve this purpose (Table 81.1).

The great majority of patients with AML fall into one of the first four WHO categories: AML with recurrent genetic changes, AML with myelodysplasia-related changes, therapy-related AML, or AML not otherwise specified (NOS) (Table 81.2).

AML with Recurrent Genetic Changes

Patients with the inv(16), t(16;16), or t(8;21) cytogenetic abnormality are lumped together into a “core-binding factor” (CBF) group. CBF refers to regulators of transcription that have a β subunit and three α subunits. RUNX1 and RUNX1T1 t(8;21) affects one of the α subunits, whereas CBFB-MYH11 affects the β subunit. Depending on the age of the population in question, CBF AML comprises 5-15% of cases and is relatively sensitive to cytosine arabinoside (ara-C), thus benefiting from “high” doses of this drug and having a relatively good prognosis.16 Despite being considered a single group, there are important clinical differences between t(8;21) and inv(16). For example, the latter is much more likely to have multiple remissions and thus longer survival.17

The t(15;17) abnormality is characteristic of FAB subtype M3, as discussed earlier. About 5% of cases of APL have normal cytogenetics, thus it is critical to assess for the PML-RARα aberration associated with the t(15;17).18 The abnormal juxtaposition of the PML and RARα genes results in the impaired promyelocytic differentiation that is the hallmark of APL. The abnormal positioning of PML leads to a characteristic pattern of nuclear immunofluorescence, demonstration of which is the quickest way to diagnose APL.19

Abnormalities in the mixed lineage leukemia (MLL) gene are associated with deletions or translocations in the long arm of chromosome 11 (11q). The clinical patterns associated with different MLL partners vary considerably. For example, pairing of MLL with MLLT3, resulting in t(9;11), leads to a prognosis better than that seen when MLL is paired with other partners.20 Thus, as with CBF, classifying an AML as “MLL AML” is itself a gross oversimplification. MLL abnormalities are particularly important to recognize because a class of drugs (DOT1L inhibitors) appear to target MLL and have produced (mainly minor) responses in clinical trials.21

The t(6;9) (which transposes the DEK and NUP214 genes) often is associated with basophilia but is perhaps mainly of interest because 80% of cases have an internal tand em duplication (ITD) of the FLT3 gene; t (6;9) is the AML subtype most frequently associated with FLT3 ITD.22 The 2008 version of WHO system does not recognize FLT3 ITD as falling in the category of recurrent genetic abnormalities (Table 81.2). This is because in perhaps 30% of cases, patients who present with an FLT3 ITD are either not FLT3 ITD positive at relapse or vice versa.23 Hence, it would appear that FLT3 ITDs are not foundational abnormalities. In contrast, a change in NPM1 or CEBPA mutation status between diagnosis and relapse is believed to be less common, leading to inclusion of these in the WHO system.24 Nonetheless, FLT3 ITD, NPM1 mutations, and CEBPA double mutations all have major prognostic importance.25NPM1 mutations and double (biallelic) CEBPA mutations convey a relatively favorable prognosis, whereas the opposite is true of FLT3 ITDs; mutations in the tyrosine kinase domain (TKD) of FLT3 are of less prognostic relevance. Of course, the significance of these aberrations depends on the context in which they occur. For example, patients in whom >50% of alleles have an FLT3 ITD have a poorer prognosis than patients with lower allelic burdens, whereas patients with NPM1 mutations and FLT3 ITDs do worse than those with only NPM1 mutations.26

It is also worth noting that patients with RUNX1-RUNX1TI [i.e., t(8;21)] or CBFB-MYH11 [i.e., inv16 or t(16;16)] are considered by WHO to have AML regardless of blast count.14 This can readily be justified on clinical grounds, given that these patients have responses to AML therapy similar to those of patients with >20% blasts.15 It is likely that blast count eventually will be considered immaterial to a diagnosis of AML in patients with NPM1 mutations and /or CEBPA double mutations.

AML with Myelodysplasia-Related Changes

Patients fall into this category if they have >=20% blasts and either morphologic features of myelodysplasia (MDS), a history of MDS, or a myeloproliferative neoplasm (MPN), or MDS-related cytogenetic abnormalities without the specific genetic abnormalities characteristic of AML, as noted in Table 81.2. MDS-related cytogenetic abnormalities are complex karyotypes (at least three distinct clonal abnormalities), monosomy of or deletion of the long arm of chromosomes 5 and /or 7 (-5,del 5q, -7, del 7q), t(3;5)(q25;q34) and changes in chromosome 11q not involving MLL.

This formulation has several problems. For example, concordance is far from perfect among pathologists in describing dysplasia. The prognostic relevance of dysplasia is in doubt.27 Finally, as noted earlier, the distinction between de novo and secondary AML (e.g., arising on a background of MDS or MPN) is probably better made based on genomics than clinical history.3

Therapy-Related AML (t-AML)

t-AMLs generally are divided into those associated with use of alkylating agents (e.g., cyclophosphamide or melphalan) and those associated with use of topoisomerase II-reactive drugs (e.g., anthracyclines or etoposide).2 The former are characterized by a 5- to 10-year latency period and complex cytogenetics. The latter more often present with abnormalities of chromosome 11q or balanced translocations, such as t(15;17) or t(8;21). Such cases generally are regarded as “AML with recurrent genetic abnormalities.”14 This is reasonable from a prognostic stand point, as cases of t-AML with t(15;17), t(8;21), or inv(16) have prognoses similar to those of cases in which these abnormalities arise seemingly spontaneously.28

Assignment of a case to the t-AML category is not always straightforward, such as when the latency period is >10 years after administration of alkylating agents. Here again, defining secondary AML based on genomic patterns likely is better than doing so based on clinical history.

AML NOS

These cases fall outside the other types of AML specified in Table 81.2. They comprise about 25% of all cases. Essentially, they are classified using FAB nomenclature. There likely will be fewer cases as more genetic subtypes of AML are recognized by the WHO.

Using data from 5,848 patients with AML NOS treated in studies by the Southwest Oncology Group (SWOG), MD and erson Cancer Center, U.K. Medical Research Council/National Cancer Research Institute (MRC/NCRI), and Dutch-Belgian Cooperative Trial Group for Hematology/Oncology and the Swiss Group for Clinical Cancer Research (HOVON/SAKK), Walter et al.29 studied the prognostic effect of different AML NOS subtypes, such as AML with minimal differentiation (M0), AML without maturation (M1), and AML with maturation (M2). After multivariate adjustment, FAB M0 was independently associated with a significantly lower likelihood of achieving complete remission and inferior relapse-free and overall survival compared with M1, M2, M4, M5, and M6, with inconclusive data regarding M7. However, when attention was restricted to known NPM1-negative patients, FAB M0 was no longer associated with worse outcomes; restricting attention to patients known to be NPM1 negative/CEPBA negative (i.e., excluding the provisional entities of “AML with mutated NPM1” and “AML with mutated CEBPA”; Table 81.2) did not affect this result. Hence, FAB subclassification of AML NOS does not provide prognostic information if data on NPM1 and CEBPA mutations are available.

81.1 Expression of cell-surface and cytoplasmic markers for the diagnosis of AML and mixed phenotype acute leukemia (MPAL)

Diagnosis of AML*
Precursor stageCD34, CD38, CD117, CD133, HLA-DR
Granulocytic markersCD13, CD15, CD16, CD33, CD65, cytoplasmic myeloperoxidase
Monocytic markersNonspecific esterase (NSE), CD11c, CD14, CD64, lysozyme, CD4, CD11b, CD36, NG2 homologue**
Megakaryocytic markersCD41 (glycoprotein IIb/IIIa), CD61 (glycoprotein IIIa), CD42 (glycoprotein 1b)
Erythroid markerCD235a (glycophorin A)
Diagnosis of MPAL***
Myeloid lineageMPO or evidence of monocytic differentiation (at least two of the following: NSE, CD11c, CD14, CD64, lysozyme)
B lineage

CD19 (strong) with at least one of the following: CD79a, cCD22, CD10

Or CD19 (weak) with at least two of the following: CD79a, cCD22, CD10

T lineagecCD3 or surface CD3

*For the diagnosis of AML, the table provides a list of selected markers rather than a mand atory marker panel.

**Most cases with 11q23 abnormalities express the NG2 homologue (encoded by CSPG4) reacting with the monoclonal antibody 7.1.

***Requirements for assigning more than one lineage to a single blast population are adopted from the WHO classification.30 Note that the requirement for assigning myeloid lineage in MPAL is more stringent than for establishing a diagnosis of AML. Note also that MPAL can be diagnosed if there are separate populations of lymphoid and myeloid blasts.

81.2 AML and related precursor neoplasms, and acute leukemias of ambiguous lineage3,30

  • Acute myeloid leukemia with recurrent genetic abnormalities
    • AML with t(8;21)(q22;q22.1); RUNX1-RUNX1T1
    • AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11
    • APL with PML-RARA*
    • AML with t(9;11)(p22.3;q23.3); MLLT3-KMT2A**
    • AML with t(6;9)(p23;q34.1); DEK-NUP214
    • AML with inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2)
    • AML (megakaryoblastic) with t(1;22)(p13.3;q13.1); RBM15-MKL1
    • AML with mutated NPM1
    • AML with biallelic mutations of CEBPA
    • Provisional entity: AML with BCR-ABL1
    • Provisional entity: AML with mutated RUNX1
  • Acute myeloid leukemia with myelodysplasia-related changes***
  • Therapy-related myeloid neoplasms****
  • Acute myeloid leukemia, not otherwise specified (NOS)
    • Acute myeloid leukemia with minimal differentiation
    • AML without maturation
    • AML with maturation
    • Acute myelomonocytic leukemia
    • Acute monoblastic/monocytic leukemia
    • Pure erythroid leukemia
    • Acute megakaryoblastic leukemia
    • Acute basophilic leukemia
    • Acute panmyelosis with myelofibrosis (syn.: acute myelofibrosis; acute myelosclerosis)
  • Myeloid sarcoma (syn.: extramedullary myeloid tumor; granulocytic sarcoma; chloroma)
  • Myeloid proliferations related to Down syndrome
    • Transient abnormal myelopoiesis (syn.: transient myeloproliferative disorder)
    • Myeloid leukemia associated with Down syndrome
  • Blastic plasmacytoid dendritic cell neoplasm
  • Acute leukemias of ambiguous lineage
    • Acute undifferentiated leukemia
    • Mixed phenotype acute leukemia with t(9;22)(q34;q11.2); BCR-ABL1*****
    • Mixed phenotype acute leukemia with t(v;11q23); KMT2A rearranged
    • Mixed phenotype acute leukemia, B/myeloid, NOS
    • Mixed phenotype acute leukemia, T/myeloid, NOS
    • Provisional entity: Natural killer (NK) cell lymphoblastic leukemia/lymphoma

Adapted from Lindsley et al.3 For a diagnosis of AML, a marrow blast count of >=20% is required, except for AML with the recurrent genetic abnormality t(15;17), t(8;21), inv(16), or t(16;16) and some cases of erythroleukemia.

*Other recurring translocations involving RARA should be reported accordingly: e.g., AML with t(11;17)(q23;q12)/ZBTB16-RARA; AML with t(11;17)(q13;q12); NUMA1-RARA; AML with t(5;17)(q35;q12); NPM1-RARA; or AML with STAT5B-RARA (the latter having a normal chromosome 17 on conventional cytogenetic analysis).

**Other translocations involving KMT2A (MLL) should be reported accordingly: e.g., AML with t(6;11)(q27;q23); MLLT4-MLL; AML with t(11;19)(q23;p13.3); MLL-MLLT1; AML with t(11;19)(q23;p13.1); MLL-ELL; AML with t(10;11)(p12;q23); MLLT10-MLL.

***More than 20% blood or marrow blasts and any of the following: previous history of MDS or myelodysplastic/myeloproliferative neoplasm (MDS/MPN), myelodysplasia-related cytogenetic abnormality (see following list); multilineage dysplasia in >=50% of cells in at least two cell lines; and absence of both prior cytotoxic therapy for unrelated disease and aforementioned recurring genetic abnormalities. Cytogenetic abnormalities sufficient to diagnose AML with myelodysplasia-related changes are:

  • complex karyotype (defined as three or more chromosomal abnormalities)
  • unbalanced changes: -7 or del(7q);del(5q) or unbalanced t(5q); i(17q) or t(17p); -13 or del(13q); del(11q); del(12p) or t(12p); idic(X)(q13)
  • balanced changes: t(11;16)(q23.3;p13.3); t(3;21)(q26.2;q22.1); t(1;3)(p36.3;q21.2); t(2;11)(p21;q23.3); t(5;12)(q32;p13.2); t(5;7)(q32;q11.2); t(5;17)(q32;p13.2); t(5;10)(q32;q21); t(3;5)(q25.3;q35.1)

****Cytotoxic agents implicated in therapy-related hematologic neoplasms: alkylating agents, ionizing radiation therapy, topoisomerase II inhibitors, and others.

*****BCR-ABL1-positive leukemia may present as MPAL but should be treated as BCR-ABL1-positive ALL.

Pathological Classification

The authors have not provided pathological classification rules.

Prognostic Factors

Prognostic Factors Required for Stage Grouping

The prognostic factors in this section are required for diagnosis of acute myeloid leukemia.

Age

Definition: chronologic

Clinical significance: Older age independently associated with more treatment related mortality(TRM) and resistance to therapy

AJCC Level of Evidence: I

Zubrod performance status(PS)

Definition: 0 or 1 = minimal symptoms; 4 = bed ridden, 3 =in bed 50-100% of time, 2= between 1 and 3

Clinical significance: PS 3-4 independently associated with TRM;

AJCC Level of Evidence: I

Hematopoietic cell transplantation comorbidity index (HCT-CI)

Definition: See Hunger et al.31

Clinical significance: Determinant of risk of TRM after hematopoietic cell transplant (HCT)

AJCC Level of Evidence: I

Cytogenetics (20 metaphase)

Definition: Categorized as “favorable”, “intermediate” or “adverse” as per Dohner et al.25

Clinical significance: Remains most important predictor of resistance to therapy; adverse and many intermediate patients are cand idates for HCT and /or trials of new therapies

AJCC Level of Evidence: I

Status of NPM,FLT3 and CEBPA genes

Definition:

a)NPM1 mutation in absence FLT3 internal tand em duplication

b) bi allelic CEBPA mutation

c) FLT3 internal tand em duplication

Clinical significance:

a)Associated with sufficiently low risk of relapse to obviate need for HCT

b) as for a)

c) Associated with sufficiently high risk of relapse as to justify HCT; patients are cand idates for combinations of chemotherapy and “FLT3” inhibitors, particularly multitargeted ones such as midostaurin

AJCC Level of Evidence: I

Additional Factors Recommended for Clinical Care

Measurable residual disease (MRD) after completion of induction or post remission chemotherapy

Definition: Assessed by multiparameter flow cytometry and /or, molecular testing for selected mutations such as NPM1 , CBFB-MYH11, RUNX1-RUNX1T1,PML-RARA

Clinical significance: Presence of MRD associated with increased probability of relapse and probably more important in ths regard than staging factors noted above

AJCC Level of Evidence: I

ASXL, TET2, and TP53 genes

Definition: Assessed for mutations

Clinical significance: Associated with sufficiently high risk of relapse to justify HCT;

AJCC Level of Evidence: I26

Clearance of mutations after induction therapy

Definition: Assessed by exome or whole genome sequencing

Clinical significance: Patients with persistence of mutations had shorter remissions than patients without persistence

AJCC Level of Evidence: III32

Bibliography

  1. Cancer Genome Atlas Research Network. Genomic and epigenomic land scapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059-2074.
  2. Churpek JE, Larson RA. The evolving challenge of therapy-related myeloid neoplasms. Best practice & research. Clinical haematology. 2013;26(4):309-317.
  3. Lindsley RC, Mar BG, Mazzola E, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood. 2015;125(9):1367-1376.
  4. Kolb EA, Meshinchi S. Acute myeloid leukemia in children and adolescents: identification of new molecular targets brings promise of new therapies. Hematology / the Education Program of the American Society of Hematology. American Society of Hematology. Education Program. 2015;2015(1):507-513.
  5. Tarlock K, Meshinchi S. Pediatric acute myeloid leukemia: biology and therapeutic implications of genomic variants. Pediatr Clin North Am. 2015;62(1):75-93.
  6. Altekruse S, Kosary C, Krapcho M, et al. SEER Cancer Statistics Review, 1975-2007, National Cancer Institute. Bethesda, MD. Fromhttp://seer.cancer.gov/csr/1975_2007/, based on November 2009 SEER datasubmission, posted to the SEER website, 2010. 2010.
  7. Kayser S, Dohner K, Krauter J, et al. The impact of therapy-related acute myeloid leukemia (AML) on outcome in 2853 adult patients with newly diagnosed AML. Blood. 2011;117(7):2137-2145.
  8. Churpek JE, Marquez R, Neistadt B, et al. Inherited mutations in cancer susceptibility genes are common among survivors of breast cancer who develop therapy-related leukemia. Cancer. 2016;122(2):304-311.
  9. Knight JA, Skol AD, Shinde A, et al. Genome-wide association study to identify novel loci associated with therapy-related myeloid leukemia susceptibility. Blood. 2009;113(22):5575-5582.
  10. Larson RA, Wang Y, Banerjee M, et al. Prevalence of the inactivating 609C-->T polymorphism in the NAD(P)H:quinone oxidoreductase (NQO1) gene in patients with primary and therapy-related myeloid leukemia. Blood. 1999;94(2):803-807.
  11. Abkowitz JL. Clone wars--the emergence of neoplastic blood-cell clones with aging. N Engl J Med. 2014;371(26):2523-2525.
  12. Welch JS, Ley TJ, Link DC, et al. The origin and evolution of mutations in acute myeloid leukemia. Cell. 2012;150(2):264-278.
  13. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. British journal of haematology. 1976;33(4):451-458.
  14. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937-951.
  15. Estey E, Thall P, Beran M, Kantarjian H, Pierce S, Keating M. Effect of diagnosis (refractory anemia with excess blasts, refractory anemia with excess blasts in transformation, or acute myeloid leukemia [AML]) on outcome of AML-type chemotherapy. Blood. 1997;90(8):2969-2977.
  16. Sinha C, Cunningham LC, Liu PP. Core Binding Factor Acute Myeloid Leukemia: New Prognostic Categories and Therapeutic Opportunities. Seminars in hematology. 2015;52(3):215-222.
  17. Marcucci G, Mrozek K, Ruppert AS, et al. Prognostic factors and outcome of core binding factor acute myeloid leukemia patients with t(8;21) differ from those of patients with inv(16): a Cancer and Leukemia Group B study. J Clin Oncol. 2005;23(24):5705-5717.
  18. Sanz MA, Grimwade D, Tallman MS, et al. Management of acute promyelocytic leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2009;113(9):1875-1891.
  19. Dyck JA, Warrell RP, Jr., Evans RM, Miller WH, Jr. Rapid diagnosis of acute promyelocytic leukemia by immunohistochemical localization of PML/RAR-alpha protein. Blood. 1995;86(3):862-867.
  20. Bernt KM, Armstrong SA. Targeting epigenetic programs in MLL-rearranged leukemias. Hematology / the Education Program of the American Society of Hematology. American Society of Hematology. Education Program. 2011;2011:354-360.
  21. Chen CW, Armstrong SA. Targeting DOT1L and HOX gene expression in MLL-rearranged leukemia and beyond. Exp Hematol. 2015;43(8):673-684.
  22. Tarlock K, Alonzo TA, Moraleda PP, et al. Acute myeloid leukaemia (AML) with t(6;9)(p23;q34) is associated with poor outcome in childhood AML regardless of FLT3-ITD status: a report from the Children's Oncology Group. British journal of haematology. 2014;166(2):254-259.
  23. Ottone T, Zaza S, Divona M, et al. Identification of emerging FLT3 ITD-positive clones during clinical remission and kinetics of disease relapse in acute myeloid leukaemia with mutated nucleophosmin. British journal of haematology. 2013;161(4):533-540.
  24. Palmisano M, Grafone T, Ottaviani E, Testoni N, Baccarani M, Martinelli G. NPM1 mutations are more stable than FLT3 mutations during the course of disease in patients with acute myeloid leukemia. Haematologica. 2007;92(9):1268-1269.
  25. Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453-474.
  26. Patel JP, Gonen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079-1089.
  27. Miesner M, Haferlach C, Bacher U, et al. Multilineage dysplasia (MLD) in acute myeloid leukemia (AML) correlates with MDS-related cytogenetic abnormalities and a prior history of MDS or MDS/MPN but has no independent prognostic relevance: a comparison of 408 cases classified as "AML not otherwise specified" (AML-NOS) or "AML with myelodysplasia-related changes" (AML-MRC). Blood. 2010;116(15):2742-2751.
  28. Quesnel B, Kantarjian H, Bjergaard JP, et al. Therapy-related acute myeloid leukemia with t(8;21), inv(16), and t(8;16): a report on 25 cases and review of the literature. J Clin Oncol. 1993;11(12):2370-2379.
  29. Walter RB, Othus M, Burnett AK, et al. Significance of FAB subclassification of "acute myeloid leukemia, NOS" in the 2008 WHO classification: analysis of 5848 newly diagnosed patients. Blood. 2013;121(13):2424-2431.
  30. Arber DA, Orazi A, Hasserjian RP. The 2016 revision to the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia. Blood. 2016.
  31. Hunger SP, Lu X, Devidas M, et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children's oncology group. J Clin Oncol. 2012;30(14):1663-1669.
  32. Moricke A, Zimmermann M, Reiter A, et al. Prognostic impact of age in children and adolescents with acute lymphoblastic leukemia: data from the trials ALL-BFM 86, 90, and 95. Klin Padiatr. 2005;217(6):310-320.