What are the therapeutic applications for CD33 modulators?

11 March 2025
Introduction to CD33 and Its Biological Role

CD33 is a member of the sialic acid–binding immunoglobulin-like lectin (Siglec) family and plays an important role in immune regulation. In the normal physiological context, CD33 is expressed on cells of the myeloid lineage such as monocytes, granulocytes, and dendritic cells, where it functions as an inhibitory receptor through its immunoreceptor tyrosine-based inhibitory motifs (ITIMs). This receptor modulates immune cell activation by dampening pro-inflammatory signals and, in certain circumstances, preventing excessive immune responses. Its structure, composed of extracellular Ig-like domains, a transmembrane region, and a cytoplasmic tail containing ITIMs, is critical in dictating its modulatory function. The receptor is capable of binding sialic acid ligands, a process that triggers downstream signaling cascades which ultimately regulate immune cell activities such as phagocytosis, cytokine secretion, and cellular proliferation.

CD33 Structure and Function

At the molecular level, CD33 is a 67 kDa single-pass transmembrane glycoprotein that exhibits two main extracellular domains: a variable (V)-set Ig-like domain that is primarily responsible for ligand binding and a constant 2-set Ig-like domain that provides structural stability. The intracellular portion contains regions that become phosphorylated to recruit tyrosine phosphatases like SHP-1 and SHP-2, thereby transmitting inhibitory signals throughout the cell. This structure is essential to its function in balancing immune responses, preventing an over-exuberant inflammatory reaction that could damage healthy tissues. Since CD33’s binding to sialylated ligands does not solely depend on its enzymatic function but also on its receptor-mediated endocytosis, research into both aspects has revealed how modulating such interactions can have wide-reaching implications in disease therapy.

Expression Patterns in Normal and Disease States

Under normal conditions, CD33 is expressed predominantly on mature myeloid cells and their precursors, demonstrating a highly regulated pattern that enables fine control of immune responses. In disease states, however, the expression of CD33 becomes dysregulated. In acute myeloid leukemia (AML) and other hematologic malignancies, for example, CD33 is overexpressed on leukemic blasts, making it a useful target for immunotherapy. Its heightened expression in malignant cells compared to normal hematopoietic stem cells and progenitors allows for selective targeting by modulators without severely affecting normal hematopoiesis. Additionally, emerging evidence suggests that in certain neurological conditions like Alzheimer’s disease, CD33 is overexpressed on microglia, negatively impacting the phagocytic clearance of amyloid plaques. The genetic association between CD33 polymorphisms and Alzheimer’s disease risk has underscored the potential of modulating CD33 function to balance neuroinflammation and promote proper clearance of protein aggregates.

Mechanism of Action of CD33 Modulators

CD33 modulators are designed to adjust the receptor’s activity, thereby influencing its downstream signaling pathways. These modulators alter CD33 function either by inhibiting its binding to ligands, enhancing its internalization, or adjusting the cellular responses following ligand engagement. The therapies can broadly be categorized by their molecular nature, ranging from monoclonal antibodies and antibody–drug conjugates (ADCs) to small molecule inhibitors, each of which exploits different aspects of CD33’s structure and function.

Types of CD33 Modulators

There are several classes of CD33 modulators investigated to date:

• Monoclonal antibodies: These are designed to bind specifically to CD33 on target cells to either induce receptor internalization or deliver cytotoxic agents through conjugation with toxic payloads. An example is gemtuzumab ozogamicin, a CD33-targeted ADC approved in the treatment of AML due to its ability to induce cell-specific toxicity.

• Bispecific antibodies: These molecules additionally recruit effector cells such as T cells by binding to CD33 on malignant cells and CD3 on T cells, which promotes targeted cellular cytotoxicity. Such bispecific T-cell engagers have shown promise in preclinical models and early stage clinical evaluations.

• CAR-T cell therapies: Although not “modulators” in the classical small molecule sense, CD33-targeting chimeric antigen receptor (CAR) T cells have been engineered to specifically recognize and eliminate CD33-positive malignant cells in AML. These cellular therapies are designed to overcome immune evasion and are being evaluated in both autologous and allogeneic settings.

• Small molecule inhibitors: In some contexts, inhibitors of CD33’s enzymatic activity or inhibitors that disrupt its interaction with sialic acid ligands have been examined. These molecules are aimed at attenuating the inhibitory signaling pathway of CD33, thereby potentially boosting immune responses in the tumor microenvironment.

Each of these modulators was developed based on detailed insights into CD33’s structure and receptor dynamics, providing multiple avenues by which the receptor’s activity can be fine-tuned for therapeutic benefit.

Biological Pathways Affected by CD33 Modulation

CD33 modulation influences several key signaling pathways. When activated, CD33 recruits phosphatases through its ITIM domains, which then dephosphorylate critical components of the immune signaling cascades. In hematologic malignancies, this inhibitory mechanism limits immune cell activation and may facilitate the survival of malignant blasts by blunting immune surveillance. Therefore, application of CD33 modulators can enhance the immune system’s ability to target leukemic cells by overcoming this suppression.

In the context of neuropathological conditions, CD33 modulates microglial activity. Increased CD33 function in microglia has been associated with inadequate phagocytosis of amyloid-β plaques in Alzheimer’s disease. By inhibiting CD33 function, modulators are able to enhance the clearance of such aggregates while balancing inflammation. Moreover, the modulation of CD33 can impact cellular processes like endocytosis and receptor recycling, which are essential for both immune regulation and neuronal homeostasis. This multifaceted impact of CD33 modulation on biological pathways makes it a unique therapeutic target with applications spanning from cancer immunotherapy to neurodegenerative disease management.

Therapeutic Applications in Disease Treatment

The therapeutic applications of CD33 modulators extend across several fields of medicine, with the most advanced research being in hematologic malignancies, while ongoing research is exploring their potential in neurological disorders. Their ability to selectively target cells overexpressing CD33 and modulate immune responses makes them attractive agents for various treatments, ranging from targeted cancer therapy to modulation of neuroinflammatory processes.

CD33 Modulators in Hematologic Malignancies

Hematologic malignancies, particularly acute myeloid leukemia (AML), have emerged as the primary therapeutic area where CD33 modulators have shown pronounced efficacy. In AML, CD33 is highly expressed on the surface of leukemic blasts, distinguishing malignant cells from normal hematopoietic stem cells. This differential expression permits targeted therapy with minimal collateral damage to normal tissues.

One of the most significant applications has been the use of CD33-targeted ADCs such as gemtuzumab ozogamicin, which became a landmark therapy. By coupling a monoclonal antibody specific for CD33 with a potent cytotoxic agent, gemtuzumab ozogamicin delivers the drug directly to CD33-positive malignant cells, leading to rapid internalization and release of the cytotoxic payload. Clinical studies have demonstrated an improvement in event-free survival when using such agents, particularly when combined with standard chemotherapy regimens in newly diagnosed AML patients. These agents have helped to lower relapse rates, improve complete remission rates, and extend overall survival in subsets of patients.

In addition to ADCs, bispecific T-cell engagers (BiTEs) have been developed to redirect T cells against CD33-positive cells. These bispecific antibodies bind to both CD33 on tumor cells and CD3 on T cells, thereby facilitating immune cell-mediated cell killing. The clinical development of bispecific CD33 modulators has been particularly promising, with early phase trials showing encouraging cytotoxicity against AML blasts even at low antigen densities. The ability to harness the patient’s own immune system to attack malignant cells is a powerful mechanism that can overcome resistance seen with traditional high-dose chemotherapies.

Furthermore, CD33-targeted CAR-T therapies have been investigated as a means to adoptively transfer engineered T cells that can recognize and destroy CD33-positive leukemic clones. Although these cellular therapies are still associated with challenges such as cytokine release syndrome and potential off-target hematologic toxicities, innovative approaches such as transient expression systems or combining CAR-T therapy with gene editing of hematopoietic stem cells (to generate CD33-deficient cells) are being explored to increase safety and efficacy. These strategies have the potential not only to improve outcomes for patients with refractory or relapsed AML but also to serve as a bridge to post-transplant consolidation therapies.

In summary, the use of CD33 modulators in hematologic malignancies is multifaceted. They enable the selective targeting of leukemic blasts through direct cytotoxicity, the engagement of immune effector cells, and even the reprogramming of cellular immunity via gene editing. These approaches have demonstrated both improved overall survival and delayed progression in clinical studies, with ongoing trials continually refining dosing regimens, reducing toxicity, and improving the therapeutic index through innovations such as allogeneic CAR-T cell products.

Potential in Neurological Disorders

Beyond hematologic applications, CD33’s role in the central nervous system has opened intriguing possibilities for neurological disorders. Genetic studies have linked certain CD33 polymorphisms with the risk of Alzheimer’s disease, suggesting an important role for CD33 in the brain’s immune surveillance mechanisms. In microglia, which are the primary innate immune cells within the central nervous system, elevated CD33 levels are associated with reduced phagocytic activity and impaired clearance of amyloid-β plaques—a hallmark of Alzheimer’s pathology. Modulating CD33 function in these cells could potentially improve the clearance of toxic aggregates and ameliorate neuroinflammation, offering a new therapeutic paradigm for neurodegenerative diseases.

Experimental studies, particularly in preclinical animal models, have shown that inhibition of CD33 signaling in microglial cells results in increased phagocytosis and reduced accumulation of amyloid-β. These findings have generated interest in the development of small molecule inhibitors and monoclonal antibodies designed to block CD33 function in the brain. Such modulators might reverse or slow the progression of Alzheimer’s disease by reactivating the microglial clearance mechanisms and restoring the delicate balance of neuroinflammatory responses. Although clinical trials focusing solely on CD33 modulators for Alzheimer’s disease are in earlier stages compared to those in oncology, the potential impact on neurodegeneration is significant, as achieving a balance between reducing amyloid load and managing inflammatory responses is critical in the disease’s management.

Additionally, CD33 modulation might indirectly influence other neurological conditions where neuroinflammation is a central component. For example, diseases with an inflammatory basis, such as multiple sclerosis or certain forms of traumatic brain injury, may benefit from precise immunomodulation. By tuning the inhibitory signals in resident immune cells, modulators of CD33 could help to mitigate pathological inflammation and promote a more regulated immune response, potentially reducing demyelination and neural damage. This therapeutic potential is still under exploration, with ongoing investigations aiming to better understand the role of CD33 in various neuropathological conditions and to optimize delivery methods that breach the blood–brain barrier while minimizing systemic effects.

Clinical Trials and Research

A significant body of research has been dedicated to evaluating the efficacy, safety, and outcomes of CD33 modulators in both hematologic malignancies and emerging neurological applications. Clinical trials conducted over the past decade have provided invaluable data that helps to illuminate both the strengths and challenges associated with these therapeutic strategies.

Current Clinical Trials Involving CD33 Modulators

For hematologic malignancies, multiple clinical trials are currently investigating various CD33-targeted therapeutic modalities. Trials evaluating the use of gemtuzumab ozogamicin in combination with induction chemotherapy in AML have reported improved survival outcomes in favorable-risk patient populations. Furthermore, bispecific antibodies and CAR-T cell therapies targeting CD33 continue to be explored in early phase trials. For instance, the PTCTC (Pediatric Transplantation and Cellular Therapy Consortium) has released promising clinical data from early phase trials using CD33CART (also referred to as VCAR33AUTO), where complete remission rates were achieved at higher dose levels in relapsed/refractory AML patients. In addition, there are studies using gene editing strategies to create CD33-deficient hematopoietic stem cells prior to transplantation, which aims to protect the normal hematopoietic compartment during subsequent CD33-targeted therapy. These approaches are part of multidimensional studies that combine cellular therapies with targeted immunotherapies to minimize off-target toxicities.

In the context of neurological disorders, while clinical trials specifically targeting CD33 in Alzheimer’s disease remain at an earlier stage, several research projects have focused on evaluating the safety and pharmacodynamics of candidate CD33 inhibitors. These trials are designed to ascertain the optimal levels of CD33 inhibition required to increase microglial uptake of amyloid-β without provoking excessive inflammation. As imaging techniques and biomarker assays become more advanced, future clinical trials in Alzheimer’s disease may incorporate biomarker endpoints such as reduction in amyloid load or improvements in cognitive function, providing a clearer picture of the therapeutic utility of CD33 modulators in neurodegenerative contexts.

Outcomes and Efficacy from Past Studies

Clinical outcomes from past studies have been instrumental in shaping current therapeutic strategies. In hematologic malignancies, gemtuzumab ozogamicin initially faced challenges related to myelosuppression and hepatic toxicity; however, modifications in dosing schedules (fractionated dosing) and improved patient selection criteria have led to re-approval and a resurgence in its clinical use. These adaptations underscore how iterative improvements in clinical practice can transform the safety profile and efficacy of CD33 modulators. Early-phase clinical trials using CD33-targeted CAR-T cells have shown rapid cytotoxic effects in AML, although longer-term persistence and potential relapse remain challenges that are driving further research into optimizing CAR design and incorporating safety-switch mechanisms.

In trials involving bispecific T-cell engagers, patient responses have been encouraging, with evidence of robust T-cell activation and elimination of leukemic blasts. These studies have demonstrated a dose-dependent relationship with tolerability profiles that improve as engineering modifications are made to minimize cytokine release syndrome and other adverse effects. In the realm of neurodegeneration, preclinical studies provide substantial evidence that reducing CD33 function in microglia enhances amyloid-β clearance, which could translate to clinical benefits if similar effects are observed in human subjects. Although clinical trial outcomes in this area are still preliminary, the trajectory of research is promising, suggesting that a fine-tuned modulation of CD33 may prevent or delay the onset of Alzheimer’s disease.

Data from these trials have also emphasized the importance of patient stratification. For instance, in AML, patients with high CD33 expression tend to benefit more from CD33-targeted therapy, highlighting the need for biomarker-driven approaches in clinical settings. This stratification is crucial not only for maximizing efficacy but also for minimizing adverse events such as off-target cytotoxicity that could damage normal hematopoietic cells. Similarly, genetic variations in CD33, such as those associated with Alzheimer’s disease risk, may eventually serve as a basis for selecting patients who might benefit from CD33 modulators, making the case for a personalized medicine approach in both oncology and neurology.

Challenges and Future Directions

While CD33 modulators have shown considerable promise, several challenges remain in both their clinical application and further development. A careful understanding of these limitations is essential to guide future research and innovation.

Limitations and Side Effects

One of the major challenges associated with CD33 modulation in hematologic malignancies is the risk of on-target, off-tumor toxicity. Because CD33 is also expressed on normal myeloid cells, therapies that target CD33 — particularly cellular therapies such as CAR-T cells — may inadvertently affect normal hematopoiesis. This can lead to complications such as prolonged myelosuppression and increased risk of infection. Early ADCs such as gemtuzumab ozogamicin faced challenges with hepatic veno-occlusive disease and other toxicities, although subsequent adjustments in dosing schedules have improved their safety profile.

In neurological disorders, achieving the appropriate degree of CD33 inhibition in the central nervous system is difficult because of the blood–brain barrier and the risk of an inflammatory response if microglial activation is excessively enhanced. While stimulating microglial phagocytosis could clear amyloid-β deposits, too much activation might trigger neurotoxic inflammatory cascades, potentially exacerbating neurodegeneration rather than ameliorating it. Furthermore, the long-term effects of modulating CD33 in the central nervous system are still not completely understood, which necessitates caution and rigorous clinical investigation before such therapies can be widely adopted.

Another limitation is the heterogeneous expression of CD33 among different patient populations. In AML, for example, the therapeutic benefit appears to be closely related to the antigen density on leukemic blasts. Patients with low levels of CD33 expression may not derive substantial benefit from CD33 modulators, which supports the need for biomarker-driven patient selection strategies. Furthermore, in the case of bispecific antibodies and CAR-T therapies, cytokine release syndrome (CRS) remains a significant adverse effect that must be managed carefully. Early trials have shown that while CRS can be transient, severe cases require prompt intervention and may limit the maximum tolerated doses of these therapeutic agents.

Future Research Directions and Innovations

Future research in CD33 modulators is likely to focus on several key areas that can enhance both the efficacy and safety of these therapies. First, there is ongoing work aimed at refining the design of antibody-based therapeutics to increase their specificity for malignant cells while sparing normal cells. This includes engineering novel linkers and payloads in ADCs to reduce off-target toxicity, or leveraging bispecific constructs that maximize immune cell recruitment yet minimize cytokine release. Advances in genetic engineering also open the possibility of creating CD33-deficient hematopoietic stem cells that can provide a safety switch during subsequent CD33-targeted immunotherapy. These approaches could allow for high-dose treatments in AML patients with reduced risk of long-term hematologic toxicity.

Another promising area lies in the optimization of CAR-T cell therapies. Refinements in CAR constructs, such as incorporating suicide genes or tuning co-stimulatory domains, are being investigated to be able to safely modulate the intensity of the T-cell response against CD33-positive malignant cells. An emerging strategy is the transient expression of CARs to reduce long-term toxicity, or the genetic knockout of CD33 in the hematopoietic compartment, which would render normal cells resistant to CAR-T mediated cytotoxicity. These strategies are being actively explored in early-phase clinical trials and have the potential to revolutionize treatment protocols for relapsed/refractory AML.

In the realm of neurological disorders, future research will likely concentrate on optimizing the delivery methods of CD33 modulators across the blood–brain barrier and defining the optimal degree of CD33 inhibition required to enhance microglial function without triggering deleterious inflammation. Developing biomarkers to monitor treatment responses in the central nervous system will be crucial. Imaging approaches such as PET scans for amyloid-β and cerebrospinal fluid biomarkers, such as neurofilament light chain concentrations, can serve as endpoints in clinical trials designed to assess the impact of CD33 modulation on disease progression.

Finally, a broader, integrative approach is required. As our understanding of cellular heterogeneity improves, efforts must be directed towards personalized medicine strategies that tailor CD33 modulating therapies to the patient’s specific profile—whether that is determined by the level of CD33 expression in AML or by the genetic variants associated with Alzheimer’s disease risk. In parallel, there is a need for innovative clinical trial designs that incorporate dynamic biomarker assessment, surrogate endpoints, and adaptive dosing strategies to accelerate the approval process while ensuring patient safety.

Conclusion

In conclusion, CD33 modulators provide a promising therapeutic avenue with a diverse range of applications across multiple diseases. The receptor’s unique biological properties, its differential expression in malignant versus normal cells, and its role in immune regulation underpin a broad spectrum of therapeutic strategies. In hematologic malignancies such as AML, CD33 modulators have advanced from ADCs and bispecific antibodies to sophisticated CAR-T cell therapies, all aimed at selectively targeting malignant blasts while sparing normal hematopoietic cells. These therapies have shown significant efficacy improvements in clinical trials, although challenges including on-target off-tumor toxicities, cytokine release syndrome, and varying antigen densities remain.

Moreover, emerging research has positioned CD33 as a potential target in neurological disorders, particularly Alzheimer’s disease. In this context, modulating CD33 to enhance microglial phagocytosis emerges as a novel approach to reducing amyloid-β deposition and neuroinflammation. Although clinical applications in this field are still in nascent stages, the groundwork laid by genetic and preclinical studies provides a robust rationale for further exploration.

Despite the significant progress, challenges such as toxicity management, patient selection, and optimizing delivery methods necessitate continued research and technological innovation. Future directions involve refining therapeutic constructs, incorporating personalized medicine strategies, and designing adaptive clinical trials that integrate dynamic biomarker endpoints to improve treatment outcomes. By addressing these challenges, the development of CD33 modulators has the potential not only to transform the management of hematologic malignancies but also to open new therapeutic pathways in neurology.

Thus, the therapeutic applications for CD33 modulators span a significant breadth of medical fields, offering general improvements in patient outcomes through specific targeting of pathogenic cells and modulation of immune pathways. The current clinical successes and cutting-edge research highlight a future where these modulators may serve as vital components of combination therapies, ultimately leading to safer and more effective treatments for complex diseases. Continued efforts in this area are expected to further unlock the potential of CD33 modulation in both oncology and neurology, paving the way for next-generation immunotherapies and neuroprotective agents that address unmet medical needs in a personalized and precise manner.

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.