Definition
Chronic myeloid leukaemia (CML) is a clonal myeloproliferative neoplasm of haematopoietic stem cell origin, characterised by dysregulated overproduction of mature and maturing granulocytes in the bone marrow and peripheral blood. This myeloid hyperplasia leads to marked leukocytosis and can be accompanied by splenomegaly.
The disease arises due to a specific chromosomal abnormality: a reciprocal translocation between chromosomes 9 and 22 [t(9;22)(q34;q11)], resulting in the formation of a shortened chromosome 22 known as the Philadelphia chromosome.
This translocation produces a fusion gene, BCR::ABL1, which encodes a constitutively active tyrosine kinase. The abnormal kinase drives the uncontrolled proliferation and prolonged survival of myeloid cells, particularly neutrophils, eosinophils, and basophils.
CML typically progresses through three clinical phases
Chronic phase
- Often asymptomatic or presenting with mild constitutional symptoms, characterised by <10% blasts in the blood or bone marrow.
Accelerated phase
- Defined by increasing blast counts (10–19%), rising basophils (>20%), persistent thrombocytopenia or thrombocytosis unresponsive to therapy, or emerging cytogenetic abnormalities.
Blast phase
- Resembles acute leukaemia, with ≥20% blasts in the blood or marrow, or the presence of extramedullary blast proliferation. This phase may manifest as either acute myeloid or acute lymphoblastic leukaemia.
Aetiology
Primary cause
- CML is a clonal myeloproliferative disorder driven by the acquired formation of the BCR::ABL1 fusion gene.
- This results from a somatic reciprocal translocation between chromosomes 9 and 22: t(9;22)(q34;q11), producing the Philadelphia chromosome.
- The fusion gene encodes a constitutively active tyrosine kinase that promotes unchecked myeloid cell proliferation.
Radiation exposure
- Ionising radiation is the only clearly established environmental risk factor.
- Risk increases with cumulative radiation dose.
Supporting evidence for radiation risk
- Atomic bomb survivors (Hiroshima and Nagasaki):
- Higher incidence of CML observed.
- Latency typically 2–10 years post-exposure.
- Greater risk noted in those exposed at a younger age.
- Occupational exposure:
- Elevated risk suggested in workers at nuclear facilities and radiologists.
- Not consistently confirmed across all studies.
- Therapeutic exposure:
- Some cases linked to prior radiation therapy, though less common.
Lack of association with other toxins
- No conclusive links to benzene, pesticides, solvents, or other environmental chemicals.
- Unlike some other leukaemias, CML does not appear to arise from such exposures.
No hereditary component
- CML does not show familial clustering.
- No known inherited genetic mutations associated with increased risk.
- Considered a sporadic disease with no identified germline predisposition.
Pathophysiology
Genetic Basis
- CML is characterised by the reciprocal translocation between chromosomes 9 and 22, resulting in the Philadelphia chromosome [t(9;22)(q34;q11)].
- This translocation fuses the BCR gene (chromosome 22) with the ABL1 gene (chromosome 9) to create the BCR::ABL1 fusion oncogene.
- The resulting BCR::ABL1 protein is a constitutively active tyrosine kinase that drives malignant transformation.
BCR::ABL1 Protein Isoforms
- The most common isoform in CML is p210, derived from fusions e13a2 or e14a2.
- p190 (e1a2): Rare in CML, more common in Ph-positive acute lymphoblastic leukaemia (ALL); associated with monocytosis and poorer prognosis.
- p230 (e19a2): Very rare; associated with indolent presentations; not routinely detected by standard PCR assays.
- Rare transcript variants include e1a3, b2a3, and e6a2.
Molecular Consequences of BCR::ABL1 Activity
- Promotes uncontrolled proliferation of myeloid progenitors.
- Impairs apoptosis via inhibition of mitochondrial cytochrome c release and caspase activation.
- Alters cell adhesion, reducing retention of progenitors in bone marrow and promoting their release into circulation.
- Causes discordant maturation, increasing immature progenitor expansion without significantly altering stem cell numbers.
Key Signalling Pathways Activated by BCR::ABL1
- MAPK pathway: Drives proliferation.
- JAK/STAT pathway: Enhances survival and proliferation.
- SAPK/JNK: Contributes to resistance to apoptosis.
- STAT5, MYC, cyclin D1: Support cell cycle progression and growth.
Disease Progression Mechanisms
- Progression from chronic phase (CP) to accelerated (AP) or blast phase (BP) involves additional genetic and epigenetic alterations.
- These changes disrupt normal differentiation and increase blast accumulation.
Contributing mechanisms include
Additional Cytogenetic Abnormalities (ACAs)
- Seen in >80% of AP/BP cases; includes:
- Trisomy 8 or 19
- Duplication of the Ph chromosome
- Isochromosome 17q, leading to TP53 loss
- Presence at diagnosis (∼7%) predicts worse response and survival.
BCR::ABL1 Activity Escalation
- Progression correlates with increased kinase activity.
- May interact with transcription factors (e.g., C/EBPα, Ikaros) to inhibit differentiation.
Mutations in Key Regulators
- TP53: Altered in 20–30% of BP patients.
- Epigenetic regulators:
- TET2, ASXL1, DNMT3A mutations associated with clonal expansion and TKI resistance.
- MYC amplification and deletions in RB1/CDKN2A are common in lymphoid BP.
- Chromosome 9 deletions: Present in 15–20% of CP cases; associated with poorer outcomes.
Epidemiology
Global Overview
- CML is a rare haematological malignancy with a global annual incidence of approximately 0.87 per 100,000 population.
- Incidence increases with age, reaching 1.52 per 100,000 in individuals over 70 years old.
- A slight male predominance is observed across populations.
- CML accounts for about 15% of all leukaemia cases in the United States.
United Kingdom
- The annual incidence of CML is approximately 1.1 per 100,000, equating to around 720 new diagnoses per year (based on 2010–2019 data).
- Peak incidence occurs in the 85–89 age group, reflecting the age-related risk increase.
United States
- In 2024, it is estimated there will be 9280 new CML cases and 1280 deaths due to the disease.
- From 2016–2020, the age-adjusted incidence rate was 1.9 per 100,000 per year, while the mortality rate was 0.3 per 100,000 per year.
- Median age at diagnosis is 66 years.
- CML is more common in males, with incidence rates of 2.2 per 100,000 in men compared to 1.4 per 100,000 in women (based on 2009–2013 data).
- Male-to-female ratio in incidence: approximately 2.5:1.5.
Trends Over Time
- In both the US and UK, incidence and mortality rates have remained relatively stable over the past decade.
- The introduction of tyrosine kinase inhibitors (TKIs) has dramatically improved survival, contributing to a growing population of patients living with CML.
Age Distribution
- While CML can occur at any age, it is primarily a disease of older adults.
- Incidence increases steadily with age, with peak diagnosis rates occurring between ages 65–89 depending on population.
History
Asymptomatic at Presentation
- Up to 50% of patients in the chronic phase are diagnosed incidentally.
- Detected through routine blood tests showing leukocytosis.
- Occasionally picked up due to splenomegaly on unrelated physical examination.
Constitutional and Non-specific Symptoms
Fatigue
- Most common complaint, especially in chronic phase (∼34%).
Weight loss
- Occurs in ∼20%; more common in accelerated or blast phases.
Loss of energy/decreased exercise tolerance
- Progressive, reflecting chronic disease burden.
Excessive sweating/night sweats
- Reported by ∼15%, typically in later phases.
Low-grade fever
- May reflect hypermetabolism or, in later stages, infection.
Shortness of breath
- Usually exertional, may reflect anaemia.
Abdominal Symptoms Related to Splenomegaly
Left upper quadrant discomfort or fullness
- Common due to splenic enlargement or infarction.
- Can refer pain to the left shoulder.
Early satiety and reduced food intake
- Due to splenic pressure on the stomach.
Left upper quadrant ‘gripping’ pain
- Suggestive of splenic infarction.
Bone and Joint Symptoms
Bone pain
- Especially in the sternum, due to bone marrow expansion.
Sternal tenderness
- Should be specifically elicited.
Arthralgia or acute gouty arthritis
- Caused by elevated uric acid from high cell turnover.
Haemorrhagic and Anaemic Symptoms
Bruising, petechiae, or epistaxis
- More frequent in accelerated or blast phases, due to thrombocytopenia or platelet dysfunction.
Pallor
- May be inferred historically via fatigue or shortness of breath, reflecting anaemia.
Retinal haemorrhages
- Rare but may be reported as blurred vision or visual disturbances.
Fever and Infection
Fever in CML
- Low-grade in chronic phase (linked to hypermetabolism).
- High-grade and associated with infections in accelerated/blast phase.
Recurrent infections
- Can indicate progression or marrow failure.
Risk Factor History
Age
- Peak incidence in US: 65–74 years (median 66 years).
- Peak in UK: 85–89 years.
- Younger median age in Southeast Asia.
Sex
- CML is more common in males (∼2.5:1.5 male:female incidence ratio).
Radiation exposure
- Prior therapeutic radiotherapy.
- Occupational exposure in nuclear industry.
- History of atomic bomb radiation (especially Japanese survivors).
Physical Examination
Splenomegaly
- Most common physical finding in CML, observed in >50% of patients at diagnosis.
- The spleen often extends >5 cm below the left costal margin.
- Size correlates with WBC count; massive splenomegaly suggests a higher disease burden.
- A rapidly enlarging spleen may herald transformation into blast crisis.
- Associated features:
- Left upper quadrant fullness or tenderness
- Referred pain to the left shoulder due to diaphragmatic irritation
- Early satiety from gastric compression
Hepatomegaly
- Occurs less frequently than splenomegaly.
- Usually due to extramedullary haematopoiesis.
- May contribute to weight loss and abdominal discomfort.
Sternal Tenderness
- May be elicited due to marrow expansion within the sternum.
- Suggestive of hyperproliferative marrow activity or progression.
Retinal Findings on Fundoscopy
- Seen in hyperleucocytic states or leukostasis.
- Examination may reveal:
- Papilloedema
- Venous congestion or obstruction
- Retinal haemorrhages
- These findings indicate hyperviscosity syndrome, often associated with very high WBC counts (>300,000–600,000/μL).
Signs of Hyperviscosity
- Physical signs may include:
- Visual disturbances
- Neurological symptoms (though these are better elicited on history)
- Fundoscopic abnormalities
- Typically seen in patients with extreme leukocytosis.
Bleeding or Bruising
- Examination may reveal:
- Petechiae
- Ecchymoses
- Mucosal bleeding (e.g. epistaxis)
- These findings are more common in the accelerated or blast phases, linked to thrombocytopenia or platelet dysfunction.
Pallor
- May be observed in patients with anaemia, due to marrow infiltration by leukaemic cells.
- Reflects disease burden or progression to accelerated/blast phase.
Soft-Tissue or Skin Infiltration
- Seen predominantly in blast phase disease.
- Physical examination may reveal:
- Skin nodules or plaques
- Lymphadenopathy (uncommon in chronic phase)
- Gingival hypertrophy or organomegaly beyond spleen/liver
Lymphadenopathy
- Rare in chronic phase.
- Presence may indicate blast crisis, especially if firm, non-tender, and generalised.
Gouty Changes (Uncommon)
- Examination may detect signs of acute gout (e.g. tender, swollen joints), usually the first MTP joint.
- Related to hyperuricaemia from rapid cell turnover.
Investigations
Initial Investigations at Diagnosis
Full Blood Count (FBC) and Differential
- Nearly all patients present with marked leukocytosis; >50% have WBC counts >100,000/μL.
- Anaemia is common (typically normocytic, normochromic).
- Platelet count may be elevated (thrombocytosis), normal, or occasionally low.
- Persistent cytopenias or cytoses unresponsive to therapy may indicate accelerated phase.
- FBC is also used for monitoring haematologic response during treatment.
Peripheral Blood Smear
- Shows a left shift with presence of:
- Myeloblasts, promyelocytes, myelocytes, metamyelocytes, and band forms
- Basophilia and eosinophilia
- Nucleated red blood cells may also be present
- Presence of a broad spectrum of myeloid precursors distinguishes CML from acute leukaemia (which exhibits a maturation arrest).
- In accelerated phase:
- Blasts ≥15%, basophils ≥20%, and promyelocytes ≥30%
- Often accompanied by thrombocytopenia and poor response to therapy
Bone Marrow Aspirate and Biopsy
- Typically reveals:
- Hypercellular marrow with marked granulocytic hyperplasia
- Increased megakaryocytes
- Mild fibrosis on reticulin staining
- Essential for:
- Confirming diagnosis
- Establishing phase (chronic, accelerated, or blast)
- Identifying additional cytogenetic abnormalities (ACAs)
- Cytogenetic analysis detects:
- Philadelphia chromosome t(9;22)(q34;q11)
- ACAs such as trisomy 8, isochromosome 17q, additional Ph chromosome, and deletions (e.g., Y chromosome)
Cytogenetic Testing
- Standard karyotyping on bone marrow cells to detect the Ph chromosome
- Recommended to analyse 20–25 metaphase cells
- Important for monitoring and detecting progression via emergence of ACAs
Fluorescence In Situ Hybridisation (FISH)
- Detects BCR::ABL1 rearrangements in interphase or metaphase cells
- Can be done on peripheral blood or bone marrow
- Used when:
- Bone marrow sample is not available
- qRT-PCR and cytogenetics yield discordant results
- Not suitable for routine monitoring if qRT-PCR is available
- Cannot detect additional cytogenetic abnormalities
Quantitative Reverse Transcriptase PCR (qRT-PCR)
- Most sensitive method for detecting BCR::ABL1 transcripts
- Should be performed:
- At diagnosis to establish baseline
- Every 3 months during treatment until BCR::ABL1 ≤1% (IS)
- Then every 3–6 months for long-term monitoring
- Can detect 1 leukaemic cell among 10⁵–10⁶ normal cells
- Used to assess molecular response, guide treatment changes, and detect minimal residual disease
Additional Investigations and Considerations
Mutational Analysis
- Indicated in:
- Inadequate response to TKI therapy
- Disease progression or relapse
- Involves next-generation sequencing (NGS) or BCR::ABL1 kinase domain mutation analysis
- Detects mutations conferring TKI resistance (e.g., T315I)
- May identify mutations in other genes (e.g., ASXL1, DNMT3A) with prognostic impact
Biochemical Tests (Metabolic Profile)
- May show:
- Hyperuricaemia due to high cell turnover
- Elevated LDH and potassium (pseudohyperkalaemia may occur)
- Useful in assessing tumour lysis risk or metabolic complications
Risk Stratification Tools
- ELTS, Sokal, or Hasford scores are calculated at diagnosis to predict prognosis and guide TKI therapy selection.
- Parameters include age, spleen size, platelet count, and peripheral blast percentage (ELTS omits eosinophils/basophils).
Imaging (Optional)
- Liver/spleen scans are rarely needed due to clear clinical findings
- Reserved for equivocal physical exams or suspected complications
- Monitor treatment response and resistance
Differential Diagnosis
Benign Reactive Conditions
Leukaemoid Reaction
- A transient response to acute infections, inflammation, or malignancy.
- Typically involves WBC counts below 50 × 10⁹/L.
- Toxic granulation, Döhle bodies, and absence of basophilia are classic features.
- Resolves with treatment of the underlying cause.
- BCR::ABL1 testing (FISH or qPCR) is negative.
Benign Neutrophilic Leukocytosis
- Seen in physiological stress, corticosteroid use, or pregnancy.
- Lacks immature granulocytes and does not show left shift.
- Investigations will show normal morphology and no clonal markers.
Other Myeloproliferative Neoplasms (MPNs)
Essential Thrombocythaemia (ET)
- Characterised by isolated and sustained thrombocytosis.
- Generally has a more indolent course and a lower risk of leukaemic transformation.
- JAK2, CALR, or MPL mutations are often found; BCR::ABL1 is absent.
- Bone marrow shows megakaryocyte proliferation without significant fibrosis.
Polycythaemia Vera (PV)
- Features elevated haemoglobin or haematocrit, often with leukocytosis and thrombocytosis.
- Almost always associated with the JAK2 V617F mutation.
- BCR::ABL1 testing is negative.
- Panmyelosis is seen in the bone marrow.
Primary Myelofibrosis (PMF)
- Typically presents with anaemia, splenomegaly, and a leukoerythroblastic blood film.
- Early PMF may resemble ET or PV.
- Bone marrow biopsy shows fibrosis, abnormal megakaryocytes, and absence of BCR::ABL1.
- Often associated with JAK2, CALR, or MPL mutations.
Chronic Neutrophilic Leukaemia (CNL)
- A rare MPN presenting with persistent neutrophilia, usually with segmented forms and minimal left shift.
- Unlike CML, there is no basophilia or monocytosis, and BCR::ABL1 is negative.
- CSF3R mutations are common.
Overlap Syndromes and MDS/MPN
Chronic Myelomonocytic Leukaemia (CMML)
- Defined by sustained monocytosis ≥1 × 10⁹/L and evidence of dysplasia.
- Peripheral smear reveals immature granulocytes and monocytes.
- Bone marrow typically shows multilineage dysplasia.
- Occasionally, PDGFR rearrangements may be identified.
- No BCR::ABL1 fusion gene is present.
Atypical CML (aCML)
- Falls under the MDS/MPN category.
- Lacks the BCR::ABL1 rearrangement and displays prominent dysgranulopoiesis and thrombocytopenia.
- Genetic studies may reveal SETBP1 or ETNK1 mutations.
Acute Leukaemias
Acute Myeloid Leukaemia (AML)
- Presents with rapid-onset symptoms, often with cytopenias, fever, or bleeding.
- Peripheral blood and bone marrow show ≥20% blasts with maturation arrest.
- Leukaemic hiatus—absence of mid-stage myeloid cells—distinguishes AML from CML.
- Cytogenetics are variable; BCR::ABL1 typically absent.
Philadelphia-Positive Acute Lymphoblastic Leukaemia (Ph+ ALL)
- A rapidly progressive B-cell lineage malignancy with systemic symptoms (fever, weight loss, sweats).
- Features anaemia, thrombocytopenia, and circulating lymphoid blasts.
- TdT+ immature B cells confirmed on flow cytometry.
- FISH/qPCR reveals p190 BCR::ABL1, in contrast to the p210 isoform seen in CML.
Management
Treatment Principles
- Primary goals: Achieve and maintain haematologic, cytogenetic, and molecular remission; prevent progression to accelerated or blast phase; and preserve quality of life.
- Secondary goals: Enable treatment-free remission (TFR) in selected patients with durable deep molecular response (DMR).
- Initial approach: TKI therapy is initiated in all patients unless contraindicated. Choice of agent is influenced by disease phase, risk stratification, comorbidities, and patient preference.
Risk Stratification
Before starting treatment, patients should be categorised using validated prognostic scoring systems:
- ELTS score (preferred): Developed during the TKI era to estimate disease-related mortality.
- Sokal score and Hasford (Euro) score: Older systems still in use, based on age, spleen size, platelet count, and blast percentages.
These scores help guide TKI selection and predict progression risk.
First-Line Therapy in Chronic Phase (CP)
Low- to Intermediate-Risk CP
- Imatinib (first-generation TKI):
- Preferred for low-risk patients and older individuals with comorbidities.
- Excellent long-term outcomes (≥80% survival at 10 years).
- Second-generation TKIs (dasatinib, nilotinib, bosutinib):
- Achieve deeper and more rapid molecular responses than imatinib.
- Preferred if TFR is a key treatment objective or if high response rates are desired early.
High-Risk CP
- Second-generation TKIs are recommended to reduce risk of progression.
- All second-generation TKIs have shown similar efficacy; choice is based on side effect profile, comorbidities, and logistics.
Asciminib
- A newer agent targeting the ABL1 myristoyl pocket.
- Suitable for patients intolerant to or failing prior TKIs.
- Active against most resistance mutations, including T315I.
- May be considered as initial therapy, particularly in cases of intolerance to other TKIs.
Monitoring and Response Milestones
Treatment efficacy is assessed by
- Complete haematologic response (CHR): Normal blood counts and spleen size.
- Cytogenetic response: Assessed via karyotyping or FISH for Ph+ cells.
- Molecular response: Measured by qRT-PCR on the international scale (IS).
Key milestones (BCR::ABL1 IS)
- 3 months: ≤10% (optimal), >10% (warning/failure)
- 6 months: ≤1% (optimal), >1% (warning), >10% (failure)
- 12 months: ≤0.1% (MMR, optimal)
Failure to achieve milestones should prompt reassessment, including adherence review, drug interaction screening, and mutational analysis.
Management of Resistance or Suboptimal Response
- Mutation testing (ABL1 kinase domain) is essential in guiding TKI switch.
- T315I mutation:
- Resistant to imatinib, nilotinib, dasatinib, bosutinib.
- Sensitive to asciminib and ponatinib.
- Multiple TKI failure:
- Consider ponatinib (third-generation TKI).
- Allogeneic haematopoietic cell transplantation (HSCT) if patient is fit and suitable donor is available.
Advanced Disease (Accelerated and Blast Phases)
- Treated with:
- A second- or third-generation TKI (based on prior use and mutational profile).
- Induction chemotherapy (AML/ALL protocols) in blast phase.
- Early donor search for HSCT is critical.
- Prognosis is significantly poorer compared to CP.
- TKI therapy may be combined with cytotoxic agents.
Alternative Therapies
- Used when TKIs are contraindicated or to control symptoms
- Hydroxyurea: Temporising measure for hyperleukocytosis or massive splenomegaly.
- Interferon alfa: Considered safe in pregnancy.
- Cytotoxic chemotherapy (e.g., cytarabine, busulfan): Historically used, rarely first-line now.
TKI Discontinuation and Treatment-Free Remission (TFR)
- Considered in patients with:
- ≥3 years on TKI
- Sustained DMR (MR4 or deeper) for ≥2 years
- No prior resistance to 2G TKIs
- Access to reliable qPCR monitoring
- Around 40–60% of patients maintain TFR; >80% if DMR sustained >5 years.
- Most patients regain DMR if relapse occurs and TKI is reintroduced.
Allogeneic HSCT
- Reserved for:
- TKI-refractory disease
- Advanced phase CML
- Multiple TKI intolerance or resistance
- Outcomes depend on:
- Phase at transplant (better in CP)
- Age and comorbidity profile
- Donor availability
- Post-transplant TKI maintenance may improve outcomes and reduce relapse risk.
Prognosis
Survival Outcomes
Historical context
- Prior to the introduction of TKIs, median survival ranged from 3 to 5 years.
- The 5-year survival rate in the early 1990s was approximately 31%.
Modern era
- SEER data (2000–2020):
- 2-year survival: 77.9%
- 5-year survival: 66.8%
- 10-year survival: 58%
- Age-adjusted 5-year survival:
- <50 years: 87.5%
- 50–64 years: 78.2%
- ≥65 years: 44.7%
- IRIS trial (imatinib):
- 89% estimated 5-year overall survival
- Only 7% progressed to accelerated or blast phase
- Second- and third-generation TKIs (e.g., dasatinib, nilotinib, bosutinib, ponatinib, asciminib) have demonstrated superior molecular responses and lower progression rates compared to imatinib.
Impact of Disease Phase
Chronic phase
- Excellent prognosis with appropriate TKI therapy.
- Long-term disease control and treatment-free remission possible in selected patients.
Accelerated phase
- Prognosis variable and depends on response to TKI therapy.
- With favourable response, survival can approximate that of chronic phase patients.
Blast phase
- Remains challenging with a poor prognosis.
- Median survival is typically 3–6 months.
- Use of TKIs combined with chemotherapy and/or haematopoietic stem cell transplantation (HSCT) may improve outcomes.
- One retrospective study reported:
- 5-year survival of 34% in patients treated with TKI and intensive therapy
- 58% in those proceeding to HSCT
Role of Comorbidities
- Comorbid conditions have become increasingly relevant due to prolonged survival.
- Charlson Comorbidity Index (CCI) has prognostic significance.
- 8-year overall survival by CCI score:
- CCI 2: 94%
- CCI 3–4: 89%
- CCI 5–6: 78%
- CCI ≥7: 46%
Prognostic Scoring Systems
Sokal Score
- Developed in the pre-TKI era.
- Uses age, spleen size, platelet count, and percentage of blood blasts.
- Categories:
- Low risk: <0.8
- Intermediate risk: 0.8–1.2
- High risk: >1.2
Hasford (Euro) Score
- Incorporates eosinophil and basophil counts in addition to Sokal parameters.
- Stratifies patients into low-, intermediate-, and high-risk groups.
- May better predict molecular response to TKI therapy.
EUTOS Score
- Simpler tool differentiating only between low- and high-risk groups.
- Based on spleen size and peripheral blood basophil count.
ELTS (EUTOS Long-Term Survival) Score
- Specifically predicts CML-related mortality in the TKI era.
- Utilises age, spleen size, platelet count, and blast percentage.
- Risk categories:
- Low risk: <1.5680
- Intermediate risk: 1.5680–2.2185
- High risk: >2.2185
- The ELTS score is now preferred for its superior prediction of CML-specific mortality.
Additional Cytogenetic Risk Factors
- Additional chromosomal abnormalities (ACAs) may emerge at diagnosis or during disease course and influence prognosis.
More favourable prognosis
- Trisomy 8
- -Y (loss of Y chromosome)
- Additional copy of Philadelphia chromosome (+Ph), if occurring in isolation
Poor prognosis (regardless of phase)
- Isochromosome 17q [i(17q)]
- Monosomy 7 or deletion 7q [-7/del(7q)]
- 3q26.2 rearrangements
High-risk combinations
- Multiple ACAs including +17, +19, +21, 11q23, or combination of +Ph with other abnormalities
- Trisomy 8 accompanied by other ACAs confers worse outcomes than trisomy 8 alone
Complications
Disease-Related Complications
Organomegaly
- Splenomegaly is common and may result in abdominal discomfort, early satiety, and in rare cases, splenic infarction.
- Hepatomegaly may occur due to extramedullary hematopoiesis.
Anaemia
- Can develop due to marrow infiltration by leukaemic cells or treatment-related myelosuppression.
Platelet Dysfunction
- Both thrombocytosis and thrombocytopenia may occur.
- Patients are at risk for bleeding and, less commonly, thrombosis.
Infections
- Increased susceptibility to bacterial and viral infections, particularly in advanced phases or during treatment with TKIs and after HSCT.
- Latent virus reactivation (e.g., varicella zoster, hepatitis B) is a concern.
- Higher severity and poor vaccination response seen in COVID-19 infections.
Constitutional Symptoms
- Bone pain and fever are more common during disease progression.
- Persistent fever may also suggest transformation to blast phase or infection.
Tyrosine Kinase Inhibitor–Associated Complications
Common Adverse Effects Across TKIs
- Myelosuppression:
- Grade 3/4 anaemia, neutropenia, or thrombocytopenia.
- Management includes dose interruption, dose reduction (~30%), and possible use of growth factors.
- Muscle Cramps and Fatigue:
- Often seen with imatinib; electrolyte replacement may be beneficial.
- Skin Reactions:
- Rash can occur with various TKIs; typically treated with corticosteroids and dose adjustment.
- QT Prolongation:
- Most concerning with nilotinib, which carries a black box warning.
- Requires ECG monitoring, avoidance of QT-prolonging drugs, and correction of electrolyte disturbances.
Agent-Specific Complications
- Dasatinib:
- Pleural Effusion: Occurs in up to 20%; may be immune-mediated. Presents with dyspnoea. Management includes TKI interruption, diuretics, corticosteroids, or switching to an alternative TKI.
- Pulmonary Arterial Hypertension: Rare (0.5–3%), presents similarly but in the absence of effusion; requires referral to pulmonary hypertension specialists.
- Ponatinib:
- Cardiovascular Events: High risk of arterial occlusion, venous thromboembolism, and heart failure. Cardiovascular assessment and dose reduction strategies are essential.
- Pancreatitis and Hepatotoxicity: Require immediate interruption and specialist input; dose reinitiation is based on risk-benefit re-evaluation.
- Posterior Reversible Encephalopathy Syndrome (PRES): Rare but serious neurological complication; symptoms include altered mental state, seizures, and vision loss.
- Nilotinib:
- Associated with vascular events and QT prolongation. Fasting administration is necessary with most formulations (unless otherwise approved). Monitoring and avoidance of interacting drugs is advised.
- Asciminib:
- Generally well tolerated but may cause pancreatitis and upper respiratory tract symptoms. Long-term safety data continue to evolve.
Haematopoietic Stem Cell Transplant–Related Complications
Infectious Risks
- Post-transplant immunosuppression increases susceptibility to opportunistic infections.
- Prophylactic antimicrobials and regular surveillance are recommended.
Graft-versus-Host Disease (GVHD)
- May occur post-transplant and contributes to long-term morbidity and mortality.
Long-Term Toxicities
- Secondary malignancies and organ dysfunction can emerge years after transplant.
References
- Baccarani M, et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia. Blood. 2013;122(6):872–884.
- Baccarani M, et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122(6):872–884.
- Branford S, et al. Predictive value of early molecular response in chronic myeloid leukemia. Blood. 2014;123(9):1353–1360.
- Breccia M, et al. Cardiovascular adverse events in chronic myeloid leukaemia patients treated with TKIs. Am J Hematol. 2019;94(9):E213–E216.
- Cortes JE, et al. Bosutinib versus imatinib in newly diagnosed chronic-phase CML. Blood. 2014;124(21):3829.
- Cortes JE, et al. Ponatinib in refractory Philadelphia chromosome–positive leukemias. N Engl J Med. 2012;367(22):2075–2088.
- Deininger MWN, Goldman JM, Melo JV. The molecular biology of chronic myeloid leukemia. Blood. 2000;96(10):3343–3356.
- Druker BJ, et al. Five-year follow-up of patients receiving imatinib for CML. N Engl J Med. 2006;355(23):2408–2417.
- Faderl S, et al. The biology of chronic myeloid leukemia. N Engl J Med. 1999;341(3):164–172.
- He W, et al. Infection risk with TKIs in CML: A population-based study. Leuk Lymphoma. 2020;61(9):2130–2137.
- Hehlmann R, et al. Impact of additional chromosomal abnormalities on survival in CML. Blood. 2016;127(24):3063–3072.
- Hehlmann R, et al. The European Treatment and Outcome Study (EUTOS) score. Blood. 2011;118(3):686–692.
- Hochhaus A, et al. ELN recommendations for treating chronic myeloid leukaemia. Leukemia. 2020;34(4):966–984.
- Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring. Am J Hematol. 2018;93(3):442–459.
- Kantarjian H, et al. Dasatinib versus imatinib in newly diagnosed chronic-phase CML. N Engl J Med. 2010;362(24):2260–2270.
- Kantarjian HM, et al. Management of myelosuppression in patients with CML receiving TKI therapy. Cancer. 2011;117(3):608–616.
- Lipton JH, et al. Asciminib in chronic myeloid leukemia after ABL kinase inhibitor failure. N Engl J Med. 2020;384(7):630–640.
- Mahon FX, et al. Discontinuation of imatinib in patients with CML with sustained complete molecular remission. Lancet Oncol. 2010;11(11):1029–1035.
- Montani D, et al. Pulmonary arterial hypertension in patients treated with dasatinib. Circulation. 2012;125(17):2128–2137.
- Nowell PC, Hungerford DA. A minute chromosome in human chronic granulocytic leukemia. Science. 1960;132(3438):1497.
- Sawyers CL. Chronic myeloid leukemia. N Engl J Med. 1999;340(17):1330–1340.
- Shah NP, et al. Dasatinib-related pleural effusion in patients with chronic myeloid leukemia. J Clin Oncol. 2008;26(25):3905–3911.
- de Lavallade H, et al. Poor anti-SARS-CoV-2 humoral and T-cell responses after COVID-19 vaccination in patients with haematologic malignancies. Br J Haematol. 2021;195(4):371–376.