section name header

Basic Information

Author: Kapil S. Meleveedu, MD

Definition

Acute lymphoblastic leukemia (ALL) is a malignancy of precursor B or T lymphocytes (lymphoblasts) characterized by uncontrolled proliferation of malignant lymphocytic cells with replacement of normal bone marrow elements and bone marrow failure. Lymphoblastic lymphoma (LBL) is diagnosed when the disease presents in extramedullary sites (most commonly as mediastinal mass in T-cell disease) and <20% of the bone marrow is involved.

Synonyms

  • Acute lymphocytic leukemia
  • Acute lymphoblastic leukemia
  • ALL
ICD-10CM CODES
C91.00Acute lymphoblastic leukemia not having achieved remission
C91.01Acute lymphoblastic leukemia, in remission
C91.02Acute lymphoblastic leukemia, in relapse
Epidemiology & Demographics

  • ALL is primarily a disease of children, adolescents, and young adults.
  • Overall incidence is 1.8/100,000 persons per year; 65% are <34 yr. It is most commonly diagnosed among people aged <20.
  • Incidence varies according to race and ethnic group, being more common in Hispanics and Whites than Blacks.
  • Male:female ratio is 55:45.

Expected overall 5 yr relative survival is 71% but substantially better in children and approaches 95%.

Physical Findings & Clinical Presentation

  • Findings consistent with bone marrow failure and peripheral cytopenias-pallor, bruising, petechiae
  • Lymphadenopathy or hepatosplenomegaly
  • Fever (disease related or infectious), bone pain, weakness, weight loss, mental status changes, and neurologic findings associated with central nervous system (CNS) involvement (if present)
  • T-cell LBL is usually associated with a mediastinal mass
  • Table E1 summarizes the clinical presentation of acute ALL and LBL

TABLE E1 Clinical Presentation of Acute Lymphoblastic Leukemia

Symptoms/SignsEtiologyManagement
FeverDisease or infectionAlways conduct fever workup and provide broad antimicrobial coverage until infectious etiology is ruled out
Fatigue, pallorAnemia (ALL infiltrating bone marrow)Packed red blood cell transfusion (slow if anemia is severe, avoid in hyperleukocytosis)
Petechiae, bruising, bleedingThrombocytopenia (ALL infiltrating bone marrow)Transfuse with platelets
PainLeukemia infiltrating bones/joints, or expanding marrow cavityEstablish diagnosis and start chemotherapy
Respiratory distress/superior vena cava syndromeMediastinal massAvoid sedation in presence of tracheal compression. Establish diagnosis as soon as possible and start chemotherapy

ALL, Acute lymphoblastic leukemia.

From Hoffman R et al: Hematology, basic principles and practice, ed 8, Philadelphia, 2023, Elsevier.

Etiology

  • Most cases are sporadic without established risk factors.
  • Ionizing radiation exposure appears to be a risk factor.
  • Down syndrome (trisomy 21) is associated with an approximately 3% risk of developing leukemia by age 30, predominantly ALL. ALL may be seen with other hereditary premalignancy syndromes (e.g., ataxia-telangiectasia).

Diagnosis

Differential Diagnosis

Disorders associated with lymphocytosis (lymphocytes >5000/mcl):

  • Adults: Chronic lymphocytic leukemia, mantle cell lymphoma, marginal zone lymphoma, hairy cell leukemia
  • Adolescents/young adults: Infectious mononucleosis syndromes due to Epstein-Barr virus or cytomegalovirus, among others, may present with lymphocyte abnormalities with appearance suggestive of leukemic blasts
  • Disorders associated with circulating blasts or blast-like cells such as acute myeloid leukemia, prolymphocytic leukemia, blastoid mantle cell lymphoma, and Burkitt lymphoma (mature B-cell leukemia/lymphoma)
  • LBL
  • Aplastic anemia; ALL may present without circulating leukemia cells and with only manifestations of bone marrow failure
Workup

  • Initial evaluation of a patient with ALL is summarized in Table E2.
  • Identification of circulating abnormal cell population by flow cytometry. CD19, cytoplasmic CD22, and/or cCD79a identifies most B lineage cells. Immature leukemic blasts should have absence of surface immunoglobulin (sIg) and will usually express CD 10, CD34, and stain positive for terminal deoxynucleotidyltransferase (TdT). A strong expression of CD20, expression of sIg, and negativity of TdT should prompt a workup for a mature B cell neoplasms such as Burkitt lymphoma because this distinction is critical. Cytoplasmic CD3 and CD7 establish immature T-cell lineage in most cases. Aberrant myeloid markers (CD13, CD33) can be seen. Early T-cell precursor (ETP-ALL) has a unique immunologic signature, as well as different prognosis and treatment approaches.
  • Cytochemical stains are sometimes easier to perform and may be available sooner but are less specific. ALL blasts should be negative for myeloperoxidase and esterase stains.
  • Bone marrow examination (Fig. E1).
  • Genetic studies define important treatment categories, of which the most important is Philadelphia chromosome positive (Ph+) vs. Philadelphia chromosome negative (Ph-) disease, because these are treated differently. Although rare in children (incidence 2% to 5%) Ph+ ALL represents the most common genetic subgroup in ALL in adults with an overall incidence of 20% to 25%. The incidence increases with age and accounts for more than 50% of cases of ALL in patients who are older than 60 yr of age.1 Ph status can be determined rapidly by polymerase chain reaction (PCR) or fluorescence in situ hybridization (FISH) and should be available within 24 to 48 h of diagnosis. The World Health Organization classification (5th edition) recognizes genetic variants of ALL as distinct syndromes (Table E3), and the clinical significance of common abnormalities is outlined in Table E4.
  • Genetic profiling for "Ph-like" ALL (genetic profile similar to Ph+ disease, but no BCR/ABL abnormality) or IKZF1 (IKAROS) mutations may provide additional prognostic information but may not be uniformly available. Ph-like ALL may respond to tyrosine kinase inhibitor therapy and may behave more like Ph+ ALL; however, Ph-like ALL cases contain a number of genetic alterations that activate kinase and cytokine receptor signaling. Alterations can be grouped into two major subclasses that include ABL-class fusions involving ABL1, ABL2, SDF1R, and PDGFRB that phenocopy BCR-ABL1, and alteration of CRLF2, JAK2, and EPOR that activate JAK/STAT signaling.
  • Other alterations in the Ph-like genomic landscape activate other kinases, including BLNK, DGKH, FGFR1, OL2RB, LYN, NTRK3, PDGFRA, PTK2B, TYK2, and the RAS signaling pathway.
  • Lumbar puncture is usually done at diagnosis, if practical, to assess for CNS involvement and to initiate CNS prophylactic therapy.

TABLE E3 WHO Classification of Precursor B-cell and T-cell Lymphoid Neoplasms

  • B-lymphoblastic leukemia/lymphoma, not otherwise specified (NOS)
  • B-lymphoblastic leukemia/lymphoma with recurrent cytogenetic abnormalities:
  • B-lymphoblastic leukemia/lymphoma with BCR::ABL1 fusion
  • B-lymphoblastic leukemia/lymphoma BCR::ABL1-like features
  • B-lymphoblastic leukemia/lymphoma with KMT2A rearrangement.
  • B-lymphoblastic leukemia/lymphoma with ETV6::RUNX1 fusion
  • B-lymphoblastic leukemia/lymphoma with ETV6::RUNX1-like features
  • B-lymphoblastic leukemia/lymphoma with high hyperdiploidy
  • B-lymphoblastic leukemia/lymphoma with hypodiploidy
  • B-lymphoblastic leukemia/lymphoma with IGH::IL3 fusion
  • B-lymphoblastic leukemia/lymphoma with TCF3::PBX-1 fusion
  • B-lymphoblastic leukemia/lymphoma with TCF3::HLF fusion
  • B lymphoblastic leukemia/lymphoma with iAMP21
  • T-lymphoblastic leukemia/lymphoma, NOS
  • Early T-precursor lymphoblastic leukemia/lymphoma

NK, Natural killer; WHO, World Health Organization.

TABLE E4 More Common Recurrent Cytogenetic Abnormalities in Acute Lymphoblastic Leukemia (ALL)/Lymphoma

AbnormalityClinical Relevance
B cell ALL
t(9;22)(q34;q11.2); BCR-ABL1Incidence approximately 3% in children, 25% in adults, rising with age; requires therapy with ABL1 tyrosine kinase inhibitors.
t(v*;11q23) KMT2A rearrangedMost common variant is t(4;11); often presents with very high white blood cell count; confers worse prognosis; rare in adults; common in infant leukemia. Hematopoietic stem cell transplant is often required.
t(12;21)(p13;q22); ETV6-RUNX1Common in children (20%-30%); rare in adults; confers improved prognosis with intensive asparaginase.
Hyperdiploidy (>50 chromosomes)Seen in about 25% of children, less in adults; confers favorable prognosis.
Hypodiploidy (<44 chromosomes)Uncommon; confers worse prognosis.
t(1;19)(q23;p13.3); TCF3-PBX1Incidence approximately 5%; intermediate/favorable in children, intermediate/poor in adults. Benefits from high dose methotrexate.
BCR-ABL1-likeShares similar gene expression profile as BCR-ABL1 but lacks the t(9;22) fusion. Adverse prognosis. Might benefit from tyrosine kinase inhibitors and/or JAK2 inhibitors.
T-cell ALLMore common in adults 60%-70%.
NOTCH1 mutation

v*, Variable gene partners. Many of these disorders also have distinct immunophenotypes by flow cytometry. Additional molecular abnormalities of recently defined relevance include mutations of IKZF1, which encodes a lymphoid transcription factor IKAROS, is associated with high relapse rates and gene expression profile similar to BCR-ABL1 translocated disease. Gene expression profiling has identified a subgroup of "Philadelphia chromosome-like" ALL with a gene expression similar to BCR-ABL1 translocation associated disease, which confers worse prognosis, but which may identify new opportunities for targeted therapies.

Figure E1 Morphologic Features of Acute Lymphoblastic Leukemia in the Blood and Bone Marrow Aspirate

A, In acute lymphoblastic leukemia (ALL), the peripheral blood count can be low, normal, or high, although frequently it is high and composed of mostly blasts. B, A similar range in cellularity is true regarding the bone marrow. Typically, lymphoblasts are small to intermediate in size and have round nuclei with dispersed chromatin and indistinct nucleoli. C, They have scant pale blue cytoplasm. A small lymphocyte (C, middle, left) is useful for comparison. D, In many cases, the lymphoblasts are monotonous, but in some cases, the lymphoblast morphology is varied with some large cells, some cells with abundant cytoplasm, and other cells with prominent nucleoli. In the older French-American-British classification, these two patterns were referred to as "ALL-L1" and "ALL-L2," respectively, although they have been shown not to have any clinical significance. Other cytologic variants of lymphoblasts are shown in E-H. These include (E) lymphocytelike blasts, (F) blasts with azurophilic granules, (G) blasts with "hand-mirror" morphology, and (H) blasts with vacuoles. The small lymphocytelike blasts can be difficult to distinguish from normal mature lymphocytes or chronic lymphocytic leukemia cells in the blood, making flow immunophenotyping important in the distinction. Lymphoblasts with granules can be misleading because they can be mistaken for myeloblast or monoblasts. The hand-mirror cells appear to be an artifact because they can be seen only focally on a smear. Vacuoles in the blasts can make the blasts difficult to distinguish from Burkitt cells. However, flow cytometry can easily distinguish the immature ALL blasts from the mature B cells seen in a leukemic presentation of Burkitt lymphoma.

(From Hoffman R et al: Hematology, basic principles and practice, ed 8, Philadelphia, 2023, Elsevier.)

TABLE E2 Initial Evaluation of a Patient with Acute Lymphoblastic Leukemia

  • Complete history (including family history)
  • Physical examination
  • CBC with differential
  • Comprehensive metabolic profile, including LFTs
  • LDH, uric acid
  • Coagulation profile
  • BM aspiration and biopsy (morphology, immunohistochemistry, flow cytometry, molecular and cytogenetic analysis)
  • HLA typing of the patient (if a potential aSCT candidate)
  • Lumbar puncture
  • Chest radiography or CT imaging of the chest

ASCT, Allogeneic stem cell transplantation; BM, bone marrow; CBC, complete blood count; CT, computed tomography; HLA, human leukocyte antigen; LDH, lactate dehydrogenase; LFT, liver function test.

From Hoffman R et al: Hematology, basic principles and practice, ed 8, Philadelphia, 2023, Elsevier.

Laboratory Tests

  • CBC reveals normochromic, normocytic anemia, thrombocytopenia.
  • Peripheral smear will usually reveal lymphoblasts, but in some cases only the marrow is involved.
  • Initial blood work should also include assessment for basic organ function (creatinine, bilirubin), blood glucose (glucocorticoids are part of therapy), and spontaneous tumor lysis syndrome (K+, Ca++, PO4++, uric acid).
  • Coagulation studies (full disseminated intravascular coagulation [DIC] screen) before lumbar puncture.
  • Studies appropriate to identifying and risk-stratifying leukemia as outlined previously.
Imaging Studies

  • Chest x-ray examination to evaluate fever and for the presence of mediastinal mass.
  • Computed tomography (CT) for symptomatic complaints. Be cautious about contrast dye exposure in patients with evidence of spontaneous tumor lysis syndrome to avoid further renal injury.

Treatment

Acute General Rx

  • Survival of children with ALL has improved from 10% to above 90% in the last 40 yr and is a major success story of modern medical science and research. Adults have fared less well, but cure rates have also improved to about 60% to 70% in standard-risk patients in recent trials. Adults in particular have benefited from tyrosine kinase inhibitor therapy for Ph+ ALL, because this disease is more common in adults and may represent 50% or more of disease in patients over 50.
  • Hyperleukocytic leukemia (white blood cell count >100,000/mcl) is uncommon in ALL and lymphocyte counts of 100,000 may be well tolerated. Prednisone and vincristine usually offer rapid cytoreduction, and leukapheresis is rarely (but sometimes) required.
  • Tumor lysis syndrome is common in ALL and was seen in 23% of patients in one large series. It is sometimes spontaneous-that is, present before therapy is given-and is a potential cause of early death. Tumor lysis syndrome is caused by release of intracellular potassium, phosphate, and nucleic acids. The nucleic acids adenosine and guanosine are eventually metabolized to uric acid. Elevated potassium may cause cardiac dysrhythmia and death. Elevated uric acid may cause renal failure through renal urate crystal deposition and possibly other mechanisms. Elevated phosphates cause renal calcium phosphate deposition and kidney injury while also lowering serum calcium, which can cause cardiac dysrhythmia and spasms. Therapy is directed mainly at maintaining renal function through vigorous hydration (3 L normal saline per day if practical, alkalinization not recommended); "forced diuresis" if necessary to maintain urine output at 2 ml/kg/h; and dialysis if necessary to control K+, phosphates, or fluid balance. Allopurinol, up to 800 mg/day for adults, 300 to 450 mg/m2/day for children, is given routinely. Rasburicase is a recombinant urate oxidase that rapidly lowers uric acid levels. The dose is 0.2 mg/kg, and one dose is usually enough. Rasburicase should be avoided in patients with G6PD deficiency. Phosphate binders are of uncertain value but are usually given. Asymptomatic hypocalcemia is not treated to avoid increasing calcium phosphate deposition. Definitions of laboratory and clinical tumor lysis and defined risk categories are outlined in Table E5.
  • Numerous protocols have been used for Ph-ALL, and the specific protocol is likely to be determined by institution/physician familiarity and access to clinical trials, among other factors.
  • In the 1990s and early 2000s, it was noted that adolescents and young adult (AYA) patients had better outcomes on pediatric trials than on adult trials. Consequently, this group (currently defined as ages 15 to 39) is now often (especially younger AYAs) treated on pediatric protocols by pediatric services or on adult "pediatric inspired" protocols.
  • Therapy for Ph-negative ALL generally has four components:
    1. Induction therapy, typically with corticosteroids, cyclophosphamide (some regimens), vincristine, an anthracycline (doxorubicin or daunorubicin usually), and asparaginase. The CD20 directed antibody rituximab has shown benefit in patients with >20% CD20 expression on their blast cells.
    2. Consolidation therapy is high-dose chemotherapy aimed at preventing relapse after remission and commonly consists of cytarabine and methotrexate in combination with other agents.
    3. Maintenance therapy is low-intensity outpatient therapy that is continued for 2 to 3 yr after completion of consolidation. Prednisone, monthly vincristine, methotrexate, and oral 6-mercaptopurine (POMP regimen) are commonly used.
    4. CNS prophylaxis is universal and is usually done with intrathecal therapy (methotrexate alone or in combination with cytarabine and hydrocortisone) administered by lumbar puncture or an Ommaya reservoir. Due to increased toxicity, cranial radiotherapy is reserved for patients with high-risk features, such as active CNS disease at diagnosis.
  • Allogeneic bone marrow transplant in first remission of ALL is controversial because of improving results with current nontransplant therapies. It is usually recommended for patients in whom the likelihood of cure is considered less than 50% to 60% with chemotherapy alone, depending on age and donor availability. Autologous bone marrow transplant is rarely used in Ph-ALL. In 2008, the final results of the MRC/UKALL XII/ECOG E2993 study were published. This study evaluated the relative safety and efficacy of chemotherapy, and autologous and allogeneic transplant after first complete remission (CR) in Ph– patients. This was a randomized trial of 1826 patients with newly diagnosed ALL. Patients who had a matched related donor were offered an allogeneic transplant. Patients without a donor were further randomized to an autologous transplant or chemotherapy and maintenance. The key conclusions of this study were:
    1. Allogeneic transplant in first CR is associated with lower relapse rates vs. autologous transplant or chemotherapy/maintenance therapy alone in Ph– patients.
    2. Allogeneic transplant in first CR improved overall survival in standard risk patients. There was a lower risk of relapse in both standard and high-risk patients. There was high treatment-related mortality in high-risk older patients. The increased mortality offsets the benefit of lower relapse risk for the patient group.
  • The findings did not support autologous transplant as replacement therapy in any ALL group.
  • More recently, treatment decisions, including the decision to proceed with allogeneic hematopoietic cell transplantation in ALL, are based on the presence or absence of measurable residual disease (MRD). MRD can be measured using multicolor flow cytometry or modalities that use next-generation sequencing (NGS) (e.g., ClonoSeQ).
  • Risk factors for treatment failure in recent protocols are outlined in Table E6. Prognostic factors in ALL are summarized in Tables E7 and E8. MRD has emerged as the strongest prognostic factor independent of traditional pretherapeutic risk factors.
  • Therapy of Ph+ ALL consists of a tyrosine kinase inhibitor-imatinib, dasatinib, nilotinib, ponatinib have been used-with chemotherapy.
    1. 2-yr survival rates are reported at 50% to 65%, with various regimens.
    2. Low-intensity induction chemotherapy with dasatinib and prednisone or imatinib, vincristine, and prednisone have resulted in remission rates of 100% and 98% and may allow for less toxicity and hospitalization at diagnosis.
    3. Allogeneic bone marrow transplant is commonly used as consolidative therapy if available but has become more controversial. Maintenance therapy with tyrosine kinase inhibitor is usually given after BMT or non-BMT therapy.
  • Therapy of relapsed disease:
    1. Allogeneic bone marrow transplant is offered for relapsed disease, but relapse after BMT is common, and long-term cure rates have been low at about 20%.
    2. In 2017, the FDA approved three new therapies for relapsed ALL: Chimeric antigen receptor T-cell (CAR-T) therapy, a form of targeted immunotherapy, yielded a remission rate of 90% in pediatric and young adult (<26 yr) patients with relapsed B-cell ALL. Chimeric antigen T-cells are created by harvesting the patient’s T cells, then transfecting them with lentivirus vector that inserts DNA expressing an anti-CD19 domain (the target antigen on B cells) coupled to a T-cell receptor. The T cells expressing the chimeric anti-CD19/T-cell receptor specifically target CD19-expressing B cells. The CAR-T cell population is then expanded ex vivo and reinfused to the patient. About 70% of remissions were durable at 6 mo. The main side effect is cytokine release syndrome (CRS) associated with "vascular leak," hypotension, respiratory and renal insufficiency, and coagulopathy. CRS is treated with tocilizumab and anti-IL6 receptor blocking antibody. CAR-T cells for ALL have been given the generic designation tisagenlecleucel (trade name Kymriah). It currently costs $475,000 and is available at centers certified for its use.
    3. In October 2021, the FDA granted approval for another CD-19 directed CART brexucabtagene autoleucel (trade name Tecartus) for treatment of adult patients (18 yr) with relapsed or refractory B-cell ALL. This was based on a single arm multicenter trial (ZUMA-3) where out of 54 patients who were evaluated for efficacy, 28 achieved CR within 3 mo. The duration of complete response was estimated to exceed 12 mo for more than half of the patients. CRS occurred in 92% of patients and neurotoxicity in 87%.
    4. Blinatumomab is a bispecific antibody that binds CD19 and CD3, redirecting T cells to leukemia cells. It was FDA-approved for relapsed ALL, including Ph+ ALL, in adults and children. Blinatumomab is administered as a continuous infusion for 4 wk (9 μg/day wk 1, 28 μg/day thereafter), with maintenance therapy for 4 wk every 12 wk. In a large phase III trial, the remission rate was 44% (vs. 25% with chemotherapy), a small number durable. In a smaller phase II trial for Ph+ ALL, the remission rate was 36%. A small number of blinatumomab responses have been durable. Blinatumomab can also be associated with cytokine release syndrome.
  • Inotuzumab ozogamicin (IO) is an antibody drug conjugate in which chemotherapeutic agent calicheamicin is bound to an anti-CD22 antibody. In a large phase III trial, the remission rate was 81% vs. 33% for standard chemotherapy, with a median duration of 4.6 mo. Approximately 40% of IO patients were successfully bridged to transplant vs. 10% with chemotherapy. A small number of responses were durable.
  • Survivorship:
    1. Survivors of childhood and adult ALL are increasingly being seen in primary care practices; as of 2006 there were estimated >50,000 survivors, likely increasing by about 2000+/yr.
    2. Long-term complications of ALL therapy include secondary malignancy from chemotherapy (usually in first 5 to 10 yr) or from radiation: If given, no plateau in risk, congestive heart failure from anthracycline therapy (often manifesting 20 to 30 yr after treatment), osteopenia and avascular necrosis from glucocorticoid therapy, obesity, and neurocognitive defects. Key recommendations include the following:
    3. Echocardiography every 3 to 5 yr for asymptomatic congestive heart failure, more often if anthracycline exposure was >250 to 300 mg/m2, because asymptomatic congestive heart failure may warrant therapy. This may show up decades after therapy.
    4. Screening for malignancy and endocrinopathies in pertinent radiation fields.
    5. Attention to the increased risk of obesity and metabolic derangement in survivors.
    6. Recent reviews (see "References") summarizing current recommendations and guidelines are accessible online (http://www.survivorshipguidelines.org/pdf/LTFUGuidelines_40.pdf).

TABLE E6 Risk Factors for Treatment Failure in Recent ALL Trials

t(v*;11q23) MLL rearranged
Hypodiploidy
Minimal residual disease after remission or consolidation*
Philadelphia chromosome-like genomic signature (in Ph-ALL)
Early precursor T (ETP) ALL (absent CD1a, CD8, weak CD5, myeloid or stem cell antigen expression)

ALL, Acute lymphoblastic leukemia.

* Measured variously after induction or consolidation therapy.

Standardized testing for this is still in development, but it may have important treatment implications. Note also that many historic risk factors (e.g., T-cell vs B-cell disease) have not been independent risk factors in current trials.

From Roberts KG et al: Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia, N Engl J Med 371:1005-1015, 2014.

TABLE E7 Prognostic Factors in Acute Lymphoblastic Leukemia

FactorPrognosisClinical Application
Age
<1 yrKMT2Ar (70%-80% infants) poor outcome; KMT2AWT same outcome as older childrenKMT2AWT do well on standard ALL therapy. Potential role for proteasome inhibitors, histone deacetylase inhibitors, hypomethylating agents, BCL2 targeted agents, immunotherapy for KMT2Ar
1-9 yrLower (standard) riskALL biology may change risk
>9 yrHigher riskALL biology may change risk
WBC
<50 × 109/LLower (standard) riskALL biology may change risk
50 × 109/LHigher riskALL biology may change risk
CNS
CNS3Higher risk of CNS and bone marrow relapseTherapy intensification, +/– cXRT
CNS2 traumatic lumbar puncture with blastsHigher risk of CNS relapseCNS directed therapy intensification
TesticularHigher riskTherapy intensification, radiation for persistent testicular leukemia
Immunophenotype
T cellHigher riskPoor outcome abolished with current therapy; nelarabine an active agent
pre-B (cIgM+)Standard riskPoor outcome abolished with current therapy
Early pre-BStandard riskGenetics may change risk
Early T-cell precursorAdverse prognosisPoor outcome abolished with current therapy
Ploidy
>50 (DI >1.16) (or trisomies of chromosomes 4 and 10)Low riskGood response to antimetabolites, may have delayed MRD clearance, but do well provided MRD <0.1% at end induction
<44Higher riskTherapy intensification, high prevalence of germline TP53 mutations
Genetic Alterations
t(9;22)/BCR-ABL1Higher riskABL TKI
t(4;11)/KMT2A-AF4Higher riskPotential role for proteasome inhibitors, histone deacetylase inhibitors, hypomethylating agents, BCL2 targeted agents, immunotherapy
t(1;19)/E2A-PBX1Higher risk of CNS relapseImproved outcome with current therapy
t(12;21)/ETV6-RUNX1Low risk
IKZF1Poor prognosis present in 80% Ph+ and also in Ph-like ALLPotential role for tyrosine kinase, JAK inhibitors
ABL-class fusionsHigh riskPotential benefits from ABL-targeting TKI
CRLF2 rearrangementsIn half of Ph-like cases, associated with Hispanic/Latino, poor outcomePotential role for JAK inhibitors
Other JAK activating lesions (JAK1/2 fusions and activating mutations, EPOR rearrangements, IL7R lesions, SH2B3 lesions)Poor prognosisPotential role for JAK inhibitors
DUX4 rearrangedFavorable prognosis. Commonly accompanied by ERG dysregulation. Frequent IKZF1 deletions, but these not associated with poor prognosis.
MEF2D fusionsPossible poor prognosis, older agePotential role for HDAC inhibition
ZNF384 fusionsYounger age, immunophenotype with weak CD10 and expression of myeloid markers; stem cell like gene expression signature; TCF3-ZNF384 fusions associated with poor prognosis
NT5C3 mutationsAssociated with 6MP/6TG resistance and relapsePossible role for NT5C3 inhibitors, inosine-5’-monophosphate dehydrogenase inhibition
CREBBPAssociated with drug resistance and relapsePotential benefit from histone deacetylase inhibitors
MRD
Day 15 <0.01%Excellent outcomeNo benefit from 2nd delayed intensification
Day 8 PB <1%Associated with excellent outcome in NCI SR patients with favorable cytogenetics, end induction MRD <0.01%Do well with relatively low-intensity therapy
Day 8 PB 1%Associated with higher risk for NCI SR patients with neutral cytogenetics even if end induction BM MRD <0.01%Benefit from intensified postinduction therapy
Slow early respondersHigher MRD=higher risk of relapse. Prognostic MRD threshold may be genotype specific (0.01% for most, but 0.1% for some favorable cytogenetic subsets)Benefit from intensified postinduction therapy
End of consolidation (or after 4 mo of therapy) >0.01%Dismal outcomeTransplantation in first CR; CD19 targeting CART being explored

ALL, Acute lymphoblastic leukemia; BM, bone marrow; CART, chimeric antigen receptor T-cell therapy; CNS, central nervous system; CR, complete remission; DI, DNA index; HDAC, histone deacetylase; JAK, Janus kinase; KMT2Ar, KMT2A-rearranged; KMT2AWT, KMT2A wild-type; MRD, minimal residual disease; NCI SR, National Cancer Institute standard risk; TKI, tyrosine kinase inhibitor; WBC, white blood cell.

Hoffman R et al: Hematology, basic principles and practice, ed 8, Philadelphia, 2023, Elsevier.

TABLE E8 Markers for Poor Prognosis in Adult Acute Lymphoblastic Leukemia

Established Risk Factors
Age>60 yr
Presenting WBC count>30,000/μL (B-cell ALL); >100,000/μL (T-cell ALL)
ImmunophenotypePro-B cell; early T cella
Cytogeneticst(4;11)(q21;q23) and other MLL rearrangements
t(9;22)(q34;q11.2) - Philadelphia chromosome
Hypodiploidy (<44 chromosomes)
Complex (>5 abnormalities)
Therapy responseTime to complete remission >4 wk
MRD0.01% at 3-6 mo after initiation of therapyb
Emerging Risk Factors
ImmunophenotypeCD20
MolecularBAALC
FUS
ERG
IKZF1c
Ph-like ALL

ALL, Acute lymphoblastic leukemia; ETP, early T-cell precursor; MRD, minimal residual disease; Ph, Philadelphia chromosome; WBC, white blood cell.

a Initial report characterizing ETP ALL showed a poor outcome. However, subsequent studies have shown variable association with response to therapy.

b Different studies have used different time points for MRD assessment.

c Focal deletions in IKZF1 are present in up to 70% of Ph-like ALL. However, IKZF1 deletions are associated with adverse outcome irrespective of association with Ph-like phenotype.

From Hoffman R et al: Hematology, basic principles and practice, ed 8, Philadelphia, 2023, Elsevier.

TABLE E5 Tumor Lysis Syndrome

Laboratory Tumor Lysis Syndromea
  • Uric acid: 8 mg/dl or 476 μmol/L
  • Potassium: 6.0 mmol/L
  • Phosphorus: 4.5 mg/dl or 1.5 mmol/L (adults), 6.5 mg/dl or 2.1 mmol (children)
    • Calcium: Correctedb Ca++ <7.0 mg/dl or 1.75 mmol/L or ionized Ca++ <1.12 mg/dl or 0.3 mmol/L, or 25% increase from baseline uric acid, potassium, phosphorus; 25% decrease for calcium
Clinical Tumor Lysis Syndrome
  • Acute kidney injury
    • Rise in serum creatinine 0.3 mg/dl (26.5 μmol/L).
    • Any creatinine >1.5 age-appropriate upper limit normal if no baseline available
    • Oliguria defined as urine output <0.5 ml/kg/h for 6 h
  • Cardiac arrhythmia
  • Seizure
  • Symptomatic hypocalcemia (e.g., neuromuscular irritability such as tetany)

a Laboratory tumor lysis syndrome present if two or more abnormalities are present within 3 days before or 7 days after therapy.

b Corrected calcium is measured calcium (mg/dl) + 0.8 × (4 - measured albumin g/dl).

From Arber DA et al: The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia, Blood 127:2391-2405, 2016.

Related Content

Suggested Readings

  1. Dekker SE : Using measurable residual disease to optimize management of AML, ALL, and chronic myeloid leukemiaAm Soc Clin Oncol Educ Book. 43:e390010, 2023.
  2. Paul S : Adult acute lymphoblastic leukemiaMayo Clin Proc. 91:1645-1666, 2016.

Related Content

  1. Foà R : Philadelphia chromosome-positive acute lymphoblastic leukemiaN Engl J Med. 386(25):2399-2411, 2022.