How do different drug classes work in treating Acute Myeloid Leukemia?

17 March 2025

Overview of Acute Myeloid Leukemia 

Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy characterized by the clonal expansion of immature myeloid progenitor cells. These cells fail to differentiate normally, leading to an accumulation of blasts in the bone marrow and peripheral blood. The pathophysiology of AML involves multiple genetic and epigenetic alterations that disrupt normal hematopoiesis, often including chromosomal aberrations, recurrent mutations (such as FLT3, NPM1, and IDH1/2) and altered gene expression patterns that promote proliferation, impaired apoptosis, and immune escape mechanisms. Modern next-generation sequencing has deepened our understanding of these molecular events and has revealed the intrinsic complexity of AML, where multiple subclones with distinct driver mutations may coexist and evolve over time. 

Current Treatment Landscape 
Historically, AML treatment was confined to a “one-size-fits-all” approach based on intensive chemotherapy with regimens such as “7+3” (cytarabine plus an anthracycline) and supportive care measures. However, owing to the diverse genetic and biological nature of this disease, treatment strategies have evolved considerably over the past decade. With the progress in genomic profiling and targeted drug discovery, the current treatment landscape not only includes conventional chemotherapy but also comprises molecularly targeted therapies and immunotherapeutic approaches. These new modalities are designed to overcome the limitations of standard treatment, reduce toxicity, and address the disease in a more personalized manner, ultimately improving outcomes for both younger and older AML patients. 

Drug Classes Used in AML Treatment 

Chemotherapy Agents 
Chemotherapy remains one of the cornerstones of AML management. The conventional chemotherapeutic regimens typically include cytarabine—which serves as an antimetabolite incorporated into DNA—and anthracyclines such as daunorubicin or idarubicin that intercalate with DNA and generate free radicals to induce cell death. These agents are designed to target rapidly dividing cells by inducing DNA damage and apoptosis. Despite their effectiveness in inducing remission, chemotherapy drugs are often associated with significant toxicity, including myelosuppression, and a substantial fraction of patients relapse because of intrinsic resistance or drug‐evading mechanisms of leukemic blasts. 

Targeted Therapies 
Targeted therapies for AML have emerged as a major new class of drugs that precisely interfere with aberrant molecular pathways driving the disease. These agents include: 
• FLT3 Inhibitors: Agents such as midostaurin and gilteritinib inhibit the FMS‐like tyrosine kinase 3 (FLT3). Since FLT3 mutations (including internal tandem duplications and point mutations) result in constitutive receptor activation, these inhibitors block the downstream signaling cascades that drive uncontrolled proliferation and survival in AML cells. 
• IDH Inhibitors: Mutations in isocitrate dehydrogenase enzymes (IDH1 and IDH2) result in the abnormal production of the oncometabolite 2-hydroxyglutarate (2-HG), which contributes to a block in cellular differentiation. Inhibitors such as enasidenib (for IDH2 mutations) and ivosidenib (for IDH1 mutations) restore normal cellular differentiation by reducing 2-HG levels. 
• BCL-2 Inhibitors: Venetoclax, a selective B-cell lymphoma 2 (BCL-2) inhibitor, induces apoptosis in AML cells by neutralizing the anti-apoptotic protein BCL-2, which is often overexpressed in leukemic blasts to protect against programmed cell death. It is commonly used in combination with hypomethylating agents or low-dose cytarabine, particularly for elderly or unfit AML patients. 
• Epigenetic Modifiers: Agents such as hypomethylating agents (azacitidine and decitabine) target aberrant DNA methylation patterns in AML. They restore normal gene expression by inhibiting DNA methyltransferases, thus promoting cell differentiation and apoptosis. In addition, inhibitors of histone deacetylases (HDAC inhibitors) and lysine demethylase 1 (LSD1) inhibitors have been investigated for their roles in reversing epigenetic silencing and inducing differentiation. 
• Other Small Molecule Inhibitors: Emerging therapies also include inhibitors targeting protein–protein interactions such as the menin–MLL interaction, which has shown promising preclinical results and is now entering clinical trials. Additionally, agents directed against other signaling partners such as PI3K/AKT/mTOR are under investigation for their potential contributions in overcoming chemoresistance. 

Immunotherapy 
Immunotherapy has revolutionized cancer treatment, and its application to AML is now an area of intense study. The immunotherapeutic strategies in AML encompass: 
• Monoclonal Antibodies (mAbs): These include unconjugated antibodies and antibody–drug conjugates (ADCs), such as gemtuzumab ozogamicin (an anti-CD33 ADC), that deliver cytotoxic payloads specifically to AML cells. They work by binding to antigens selectively expressed on leukemic blasts and triggering immune-mediated killing as well as direct cytotoxicity. 
• Checkpoint Inhibitors: These agents block inhibitory pathways (e.g., PD-1/PD-L1, CTLA-4) to reinvigorate T-cell responses against AML. Although success in solid tumors has driven interest, responses in AML have been variable due to factors such as low mutational burdens and the immunosuppressive bone marrow microenvironment. 
• Chimeric Antigen Receptor (CAR) T- and NK-cell Therapies: These novel approaches genetically modify a patient’s immune effector cells to express receptors targeting specific AML-associated antigens (such as CD123, CD33, and FLT3). The modified cells are then reinfused to seek and destroy leukemic cells. While promising, their development in AML is challenged by the lack of truly tumor-specific antigens and potential on-target off-tumor toxicities. 
• Vaccination Strategies: Peptide or dendritic cell-based vaccines aim to stimulate the patient’s endogenous immune response against leukemic antigens. Although these strategies have shown limited efficacy on their own, they may be useful when combined with other immunomodulatory approaches. 

Mechanisms of Action 

How Chemotherapy Works 
Chemotherapeutic agents used in AML primarily target rapidly proliferating leukemic cells by interfering with DNA synthesis and cell division. Cytarabine, a cytosine analog, is metabolized intracellularly and incorporated into DNA during the S-phase of the cell cycle, leading to chain termination and subsequent apoptosis. Anthracyclines, such as daunorubicin and idarubicin, intercalate between DNA base pairs, disrupting topoisomerase II–mediated DNA repair mechanisms, and generate reactive oxygen species that induce additional DNA damage. Together, these drugs cause a cytotoxic effect that reduces the tumor burden. However, their mechanism is non-selective, affecting not only AML blasts but also normal hematopoietic stem cells, thereby accounting for the side effects such as myelosuppression. 

Mechanisms of Targeted Therapies 
Targeted therapies work by interfering with specific molecular pathways that are aberrantly activated in AML cells. Their mechanisms include: 
• Inhibition of Oncogenic Signaling: FLT3 inhibitors act by binding to the active conformation of the mutated receptor tyrosine kinase FLT3, thereby inhibiting downstream signaling pathways (including PI3K/AKT, RAS/MEK/ERK, and STAT5) that are crucial for cell proliferation and survival. In doing so, these agents curtail the uncontrolled cell growth characteristic of FLT3-mutated AML. 
• Restoration of Differentiation: The IDH inhibitors block the neomorphic enzymatic activity of mutant IDH enzymes, leading to a reduction in the oncometabolite 2-HG. This alleviation of metabolic block restores normal epigenetic regulation and allows the leukemic blasts to resume terminal differentiation. 
• Promotion of Apoptosis: Venetoclax, the BCL-2 inhibitor, disrupts the balance between pro-apoptotic and anti-apoptotic proteins in AML cells. By inhibiting BCL-2, which normally sequesters pro-apoptotic factors, venetoclax triggers the intrinsic apoptosis pathway leading to cell death. This mechanism is particularly effective in cells that rely heavily on BCL-2 for survival. 
• Epigenetic Reprogramming: Hypomethylating agents (azacitidine and decitabine) incorporate into DNA and inhibit DNA methyltransferases, causing hypomethylation and reactivation of silenced genes, including tumor suppressor genes. HDAC inhibitors interfere with histone deacetylation, leading to a more open chromatin structure that reactivates gene expression programs favoring differentiation and apoptosis. 
• Disruption of Protein–Protein Interactions: Novel agents such as menin–MLL inhibitors block interactions that are critical for maintaining the leukemic state, thereby helping the cells to undergo differentiation and lose their self-renewal capacity. This targeted interruption of oncogenic protein complexes is a promising area of research. 

Immunotherapy Mechanisms 
Immunotherapy leverages the host’s immune system to recognize and eradicate AML cells. The mechanisms include: 
• Direct Targeting via Monoclonal Antibodies: mAbs bind to surface antigens (such as CD33 or CD123) on AML blasts. Once bound, these antibodies can induce cell death through mechanisms like antibody-dependent cellular cytotoxicity (ADCC), complement-mediated cytolysis, or direct induction of apoptosis through receptor modulation. ADCs add another layer by delivering a cytotoxic payload directly to the cells upon internalization, thereby amplifying the killing effect while limiting systemic toxicity. 
• Checkpoint Blockade: By inhibiting checkpoints such as PD-1 or CTLA-4, checkpoint inhibitors release the brakes on T cells, allowing them to recognize and attack AML cells more effectively. This strategy aims to overcome the immune suppressive signals within the leukemic bone marrow microenvironment. 
• Adoptive Cell Therapies: CAR-T and CAR-NK therapies are designed to reprogram a patient’s immune cells to express engineered receptors that specifically target AML-associated antigens. These receptors guide the modified immune cells to the malignant cells, resulting in targeted lysis. Although promising, the overlapping expression of target antigens on normal cells remains a challenge. 
• Vaccination Approaches: By presenting AML-specific antigens via peptide vaccines or dendritic cell vaccines, the immune system can be primed to recognize and mount a response against leukemic cells. While these vaccines induce antigen-specific T-cell responses, their clinical efficacy has so far been limited, necessitating further research and combination with other therapies. 

Clinical Efficacy and Outcomes 

Comparative Effectiveness 
The comparative effectiveness of various drug classes in AML is influenced by disease subtype, patient age, mutational profile, and treatment setting (e.g., newly diagnosed versus relapsed/refractory AML). Conventional chemotherapy, while effective in inducing remission in a significant percentage of younger patients, results in high relapse rates and considerable toxicity. In contrast, targeted therapies generally offer a more favorable toxicity profile and can be used in patients who are not candidates for intensive chemotherapy. For example, venetoclax-based regimens, when combined with hypomethylating agents, have significantly improved the overall survival and quality of life in elderly AML patients, even though resistance remains a challenge. Immunotherapy, particularly monoclonal antibodies like gemtuzumab ozogamicin, has shown efficacy in specific molecular subtypes of AML and when used in combination with chemotherapy, offers improved outcomes by reducing minimal residual disease and the risk of relapse. Comparative studies and meta-analyses have demonstrated that while no single therapy is universally superior, treatments tailored to the patient’s genetic and clinical profile can yield the best results. For instance, patients with FLT3 mutations benefit particularly from FLT3 inhibitors, whereas those with IDH mutations respond well to IDH inhibitors. 

Case Studies and Clinical Trials 
Numerous clinical trials and case studies have advanced our understanding of AML treatment using different drug classes. Early-phase trials with FLT3 inhibitors like midostaurin and gilteritinib have demonstrated improved complete remission rates and overall survival in FLT3-mutated AML patients. Similarly, phase II studies of venetoclax in combination with azacitidine or decitabine have resulted in high response rates and manageable toxicity profiles in elderly patients or those ineligible for intensive chemotherapy. ADCs such as gemtuzumab ozogamicin have undergone several clinical trials that have led to their approval in selected AML populations based on favorable outcomes in reducing relapses, particularly in core-binding factor AML. In the area of immunotherapy, early-phase trials with CAR-T cells targeted against antigens such as CD33 and CD123 have shown potent preclinical efficacy, although translation into clinical practice has been challenged by issues related to antigen specificity and toxicity. These case studies underscore that clinical efficacy is highly dependent on the alignment of drug mechanism with the underlying molecular pathology of the disease. 

Challenges and Future Directions 

Resistance Mechanisms 
One of the most significant challenges in treating AML is the development of drug resistance. Resistance mechanisms are multifactorial and include: 
• Intrinsic Resistance: AML cells inherently have genetic and epigenetic polymorphisms that confer survival advantages. For example, overexpression of anti-apoptotic proteins such as MCL-1 can render BCL-2 inhibitors like venetoclax less effective, necessitating the use of combination therapies to overcome this resistance. 
• Acquired Resistance: Clonal evolution under the selective pressure of therapy can result in the emergence of new subclones with additional mutations. Alterations in key signaling pathways, such as secondary mutations in FLT3 during treatment with FLT3 inhibitors, further contribute to the loss of therapeutic efficacy. 
• Microenvironment-Mediated Resistance: The bone marrow niche provides a protective environment by secreting cytokines and growth factors that support AML cell survival and reduce the effectiveness of chemotherapy and targeted agents. This includes altered immune checkpoints that contribute to an immunosuppressive milieu. 
These resistance mechanisms emphasize the need to monitor minimal residual disease (MRD) and clonal evolution closely during and after treatment, using advanced techniques such as sequencing and flow cytometry, to adapt therapeutic strategies in a timely manner. 

Emerging Therapies and Research 
Research into novel therapeutic approaches in AML is focusing on several areas: 
• Combination Therapies: One promising direction is the use of synergistic combinations of drug classes. For instance, combining venetoclax with hypomethylating agents has shown promise in overcoming resistance and achieving deeper, longer-lasting remissions. Similarly, combining targeted therapies with immune checkpoint inhibitors may enhance the overall antileukemic immune response. 
• Next-Generation Targeted Agents: New targets such as the menin–MLL interaction, epigenetic regulators, and other protein–protein interactions are being explored. Clinical trials with agents that disrupt these interactions have shown promising preclinical data, and further research is underway to establish their clinical efficacy. 
• Adoptive Cell Therapies: Advances in CAR-T and CAR-NK cell technologies continue to progress. Ongoing trials are optimizing antigen selection and cell engineering techniques to mitigate off-tumor toxicities and enhance the persistence and efficacy of the modified immune cells. Research is also focusing on overcoming the barriers posed by antigen escape and the immunosuppressive bone marrow environment. 
• Biomarker-Driven Approaches: Enhanced molecular profiling is increasingly enabling personalized treatment strategies. The integration of genomic, transcriptomic, and immunophenotypic data into risk stratification models is being used to tailor treatments to individual patients, thereby potentially improving therapeutic outcomes. 
• Overcoming Drug Resistance: Investigational drug combinations and agents targeting resistance pathways—such as those that modulate apoptosis regulators like MCL-1—are actively being studied. Future research is emphasizing early detection of resistance markers using advanced diagnostics with the aim of preemptively adapting treatment regimens. 

In summary, the future of AML treatment lies in a multi-pronged strategy: deploying combination therapies, leveraging new insights into drug resistance, and applying state-of-the-art technologies for patient stratification and MRD monitoring. These efforts pave the road toward personalized treatments with the ultimate goal of transforming AML into a manageable condition with long-term survival. 

Detailed Conclusion 
The treatment of acute myeloid leukemia today involves a diverse array of drug classes that work through multiple mechanisms. Conventional chemotherapy, despite its non-specific approach and considerable toxicity profile, forms the foundation of AML management by directly damaging the DNA of rapidly dividing cells, resulting in remission induction. However, its limitations—including high relapse rates and significant side effects—have driven the development of targeted therapies. These newer drugs, such as FLT3 inhibitors, IDH inhibitors, BCL-2 inhibitors, and epigenetic modifiers, work by selectively blocking the aberrant signaling pathways that underlie leukemogenesis. Through the inhibition of oncoproteins, restoration of normal cell differentiation, and promotion of apoptosis, targeted therapies address the genetically diverse landscape of AML, offering a more personalized therapeutic approach. 

Immunotherapy further expands the treatment armamentarium by harnessing the immune system. Monoclonal antibodies, checkpoint inhibitors, and adoptive cell therapies such as CAR-T cells act by enhancing the immune recognition and elimination of AML cells. Although these strategies are promising, challenges such as on-target off-tumor toxicity and an immunosuppressive bone marrow microenvironment must be overcome. Combining these therapies with conventional chemotherapy or with one another may provide synergistic effects and help overcome resistance mechanisms that curtail the efficacy of single-agent treatments. 

The clinical efficacy of these drug classes has been evaluated in various trials and case studies, which have demonstrated that the integration of targeted therapies and immunotherapies into the standard treatment protocols can improve overall survival, complete remission rates, and quality of life for patients. However, the issue of drug resistance—both intrinsic and acquired—remains a significant barrier that demands continuous research and innovation. Resistance mechanisms, such as clonal evolution or microenvironment-mediated protection, underscore the complexity of AML and the need for iterative or combination treatment strategies that are adaptable over time. 

Looking toward the future, emerging research is focused on better understanding the molecular drivers of AML, developing next-generation targeted agents, and refining immunotherapeutic approaches. The integration of advanced biomarker analyses and dynamic MRD monitoring into clinical practice is expected to facilitate early intervention when resistance begins to develop. Collectively, this new paradigm of personalized, multi-targeted, and adaptive therapy holds substantial promise for overcoming the limitations of current treatments, ultimately aiming to transform AML from an acutely life-threatening condition into a chronic disease that can be managed with long-term remission and improved survival outcomes. 

In conclusion, while the drug classes used in AML treatment—ranging from conventional chemotherapy and targeted therapies to novel immunotherapies—operate through distinct mechanisms, they share the common goal of eliminating leukemic cells and enhancing patient survival. The general approach begins with conventional therapies to induce remission, followed by tailored interventions based on the patient’s genetic profile and disease dynamics. This general-to-specific-to-general strategy, wherein initial broad application is refined by increasingly specific targeted measures, represents the evolving treatment paradigm in AML. The success thus far, as evidenced by improved outcomes in clinical trials and real-world studies, confirms that harnessing multiple perspectives—from molecular biology and pharmacology to immunology—is key to the future of AML therapy. Continued research into resistance mechanisms and the development of innovative combination treatments will be essential to fully realize the potential of these diverse drug classes. Ultimately, the integration of precision medicine and immunotherapy into AML treatment protocols promises to usher in an era of improved outcomes and personalized care for patients battling this devastating disease.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.