Chronic Myelomonocytic Leukemia: The Dual-Natured Blood Cancer

More Than Just Monocytes

Imagine a disease that simultaneously possesses the characteristics of two different types of blood cancers, defying easy classification and challenging treatment paradigms. This is chronic myelomonocytic leukemia (CMML), a rare but aggressive hematologic malignancy that walks the line between myelodysplastic syndromes (ineffective blood cell production) and myeloproliferative neoplasms (excessive blood cell production). With an incidence of less than 1 case per 100,000 people annually, CMML is considered a rare disease, yet its impact is profound 1 .

Incidence

<1 per 100,000 annually

AML Transformation

15-20% within 3-5 years

Median Age

Over 70 years

Genetic Mutations

>95% of patients

The clinical journey of CMML patients is particularly challenging—approximately 15-20% will transform to acute myeloid leukemia (AML) within 3-5 years of diagnosis, making it one of the most unpredictable myeloid malignancies 4 7 . What makes CMML especially relevant in today's aging society is its striking predilection for older adults, with a median age at diagnosis of over 70 years 4 .

Understanding CMML: A Disease of Two Faces

Myelodysplastic CMML (MD-CMML)
  • White blood cell counts <13,000/μL
  • Primary problem: Hematopoietic insufficiency
  • Common symptoms: Cytopenias (anemia, low platelets)
  • Clinical resemblance: Myelodysplastic syndromes
  • Common genetic mutations: Epigenetic regulators, spliceosomes
Myeloproliferative CMML (MP-CMML)
  • White blood cell counts ≥13,000/μL
  • Primary problem: Excessive cell production
  • Common symptoms: Splenomegaly, constitutional symptoms
  • Clinical resemblance: Myeloproliferative neoplasms
  • Common genetic mutations: RAS pathway mutations

What Exactly is CMML?

Chronic myelomonocytic leukemia is a clonal hematopoietic stem cell disorder characterized by sustained peripheral blood monocytosis (increased monocyte counts) and bone marrow dysplasia (abnormal development of blood cells) 1 7 . The World Health Organization (WHO) and International Consensus Classification (ICC) have established specific diagnostic criteria, including:

  • Sustained peripheral blood monocytosis (≥0.5 × 10⁹/L for over 3 months)
  • Monocytes comprising ≥10% of the white blood cell count
  • Presence of dysplasia in one or more myeloid lineages
  • Less than 20% blasts (immature cells) in blood and bone marrow
  • Evidence of clonality through cytogenetic or molecular testing 1 7

The Molecular Landscape: Unraveling CMML's Genetic Blueprint

The pathogenesis of CMML is deeply rooted in age-related clonal hematopoiesis, where hematopoietic stem cells accumulate mutations over time 4 . By age 70, most blood cells are derived from only 10-20 stem cell clones, some of which carry cancer-driving mutations 4 . More than 95% of CMML patients harbor somatic mutations, with specific patterns influencing disease behavior and prognosis 1 7 .

Epigenetic Regulators

∼60-90% of cases

Mutations in genes like TET2 (∼60%), ASXL1 (∼40%), and DNMT3A disrupt normal chemical modifications that control gene expression.

Spliceosome Components

∼50% of cases

Mutations in SRSF2 affect how messenger RNA is processed, leading to abnormal protein production.

Signaling Pathways

∼30% of cases

Mutations in RAS pathway genes (NRAS, KRAS, CBL, NF1) are associated with proliferative phenotype.

Key Genetic Mutations in CMML

Gene Frequency Functional Category Clinical Significance
TET2 ∼60% Epigenetic regulator Often ancestral mutation; better prognosis when sole mutation
SRSF2 ∼50% RNA splicing Frequently co-occurs with TET2 mutations
ASXL1 ∼40% Chromatin modification Truncating mutations predict inferior outcomes
RAS Pathway ∼30% Cell signaling Associated with proliferative phenotype and AML transformation

Spotting the Enemy: Diagnostic Approaches for CMML

Diagnosing CMML requires a multifaceted approach that goes beyond simple blood counts. The current diagnostic workflow includes:

Complete Blood Count with Differential

Reveals persistent monocytosis (≥0.5 × 10⁹/L) and may show other abnormalities like anemia or thrombocytopenia 1 7 .

Bone Marrow Examination

Assesses cellularity, dysplasia, fibrosis, and blast percentage (must be <20%) 1 .

Cytogenetic Analysis

Identifies chromosomal abnormalities present in approximately 30% of patients 7 .

Molecular Genetic Testing

Next-generation sequencing panels detect somatic mutations in the characteristic CMML genes, providing both diagnostic and prognostic information 1 7 .

Flow Cytometry

Can identify abnormal partitioning of monocyte subsets and detect immature granulocytes with immunosuppressive properties 1 3 .

A Closer Look: Tracking Immature Granulocytes Through Flow Cytometry

Recent research has highlighted the importance of immature granulocytes (iGRANs) in CMML progression. These cells not only belong to the malignant clone but also function as immunosuppressive cells that promote disease progression by suppressing the immune system's ability to fight the cancer 3 . A 2025 study published detailed a sophisticated flow cytometry protocol to quantify these cells, providing clinicians with both diagnostic and prognostic tools 3 .

Methodology: Step-by-Step Protocol
  1. Sample Collection: 5-10 mL of peripheral blood is collected in EDTA tubes and processed within 24 hours to maintain cell viability 3 .
  2. PBMC Isolation: Peripheral blood mononuclear cells are separated using Ficoll gradient centrifugation 3 .
  3. Cell Staining: The PBMCs are labeled with a panel of fluorescent antibodies targeting specific surface markers 3 .
  4. Flow Cytometry Analysis: The stained cells are analyzed using a flow cytometer 3 .
Results and Significance

This protocol allows for both relative quantification (iGRANs as a fraction of myeloid cells) and absolute quantification (absolute iGRAN count per volume of blood) 3 . The accumulation of these immunosuppressive cells in peripheral blood correlates with poor outcomes, similar to observations in solid tumors and severe sepsis 3 . This method provides clinicians with a valuable tool for patient stratification and monitoring treatment response.

Essential Research Reagents
Reagent Specific Example Function in Experiment
Antibody Panel APC anti-human CD56, FITC anti-human CD66b, Percp5.5 anti-human CD11b, etc. Cell surface staining for population identification
Density Gradient Medium Ficoll solution (density 1.077 g/mL) Separation of mononuclear cells from whole blood
Staining Buffer PBS with fetal bovine serum Provides medium for antibody staining
Hemolytic Solution Ammonium chloride-based Lyses residual red blood cells after separation
Flow Cytometer Fortessa cytometer (5 lasers) Cell analysis and data acquisition

Current Treatment Landscape: Limited Options, Growing Hope

The treatment arsenal for CMML remains limited, with approaches tailored to individual patient factors including disease subtype, risk stratification, and eligibility for intensive therapy.

Established Treatments
Allogeneic Stem Cell Transplantation

The only potentially curative option for CMML, but limited by patient age and comorbidities given the typical diagnosis age >70 years 1 7 .

Hypomethylating Agents (HMAs)

Azacitidine and decitabine are the only FDA-approved drug therapies for CMML. These drugs work by reversing abnormal DNA methylation patterns, potentially reprogramming cancer cells 6 7 . However, their effectiveness is limited, particularly in proliferative subtypes with RAS mutations 6 .

Hydroxyurea

A chemotherapy drug that can help control blood counts, particularly in proliferative CMML, and may extend survival for some patients 6 7 .

Emerging and Investigational Therapies

The CMML therapeutic pipeline is remarkably active, with over 20 companies developing more than 25 investigative candidates 2 . Promising approaches include:

Immunotherapy

IO-202, a first-in-class monoclonal antibody targeting LILRB4 (a receptor highly expressed on CMML cells), has received FDA fast-track designation and is in Phase I/II trials 6 .

Novel Targeted Agents

Timdarpacept, a recombinant SIRPα fusion protein that blocks the "don't eat me" signal to immune cells, is in Phase III development 2 .

Combination Therapies

The PREACH-M trial investigating azacitidine with lenzilumab (an antibody against GM-CSF) has shown remarkable early results 5 .

The PREACH-M Trial: A Beacon of Hope

The PREACH-M trial represents one of the most promising recent developments in CMML treatment. This Australian study, led by Associate Professor Dan Thomas, combines the standard therapy azacitidine with the investigational antibody lenzilumab 5 .

Groundbreaking Results

Interim results published in Blood journal demonstrated 5 :

  • 22 of 34 participants showed significant improvements within the first year
  • Harmful cells and inflammatory markers dropped dramatically after three months
  • Benefits were sustained, with 15 patients continuing treatment beyond one year without relapse
  • Three participants have remained on therapy for over three years
  • One patient maintains remission after four years of treatment

Associate Professor Thomas captured the significance of these findings: "Not only are patients experiencing short-term improvements in their blood counts and symptoms; they're maintaining those improvements for years. That's something we haven't been able to achieve before" 5 .

The success of this combination therapy suggests that simultaneously targeting the malignant clone (with azacitidine) and the inflammatory microenvironment (with lenzilumab) may provide synergistic benefits for CMML patients.

Future Directions and Conclusion

The landscape of CMML research and treatment is rapidly evolving. Beyond the therapies mentioned, investigators are exploring CAR-T cell therapies, JAK inhibitors, and various combination regimens to improve outcomes 8 . The global CMML market is poised for significant growth as these innovative approaches mature, driven by increasing disease recognition in aging populations and advances in personalized medicine 8 .

Digital Health Technologies

The integration of digital health technologies, such as remote monitoring and telemedicine, may further enhance long-term disease management for CMML patients 8 .

Companion Diagnostics

The development of companion diagnostics like liquid biopsies and next-generation sequencing is enabling more accurate diagnosis and personalized treatment plans 8 .

In conclusion, while CMML remains a challenging dual-natured malignancy, the growing understanding of its molecular foundations, combined with innovative therapeutic strategies, is transforming the outlook for patients. From sophisticated diagnostic tools like iGRAN quantification to groundbreaking clinical trials like PREACH-M, the future of CMML management is increasingly promising, offering hope for more effective and targeted treatments in the years to come.

References

References