What are the new molecules for CD33 modulators?

11 March 2025
Overview of CD33 and Its Biological Role

CD33, also known as Siglec-3, is a transmembrane receptor of the immunoglobulin superfamily that plays an important role in the regulation of immune responses. It is expressed mainly on cells of the myeloid lineage, including monocytes, granulocytes, dendritic cells, and microglia, and is implicated in both innate and adaptive immune functions. Detailed studies have illuminated its structure, isoform diversity, and contributions to immunosuppressive signaling pathways. Overall, CD33 is a critical molecule that influences both inflammation and cellular homeostasis, with its modulation representing an important therapeutic strategy for disorders such as acute myeloid leukemia (AML) as well as neurodegenerative conditions like Alzheimer’s disease.

CD33 Structure and Function

At the molecular level, CD33 appears as a 67 kDa single-pass transmembrane glycoprotein. Its extracellular region is composed of two immunoglobulin-like (Ig) domains: an N-terminal V-set domain which is primarily responsible for binding sialic acid and a subsequent C2-set domain. The V-set domain is crucial for epitope recognition by most existing therapeutics, whereas the C2-set domain is located closer to the cell membrane and is emerging as a promising alternative target. CD33’s cytoplasmic domain contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs), which, upon phosphorylation, recruit phosphatases like SHP-1 and SHP-2—key mediators of inhibitory signaling. In addition to its principal full-length isoform (CD33FL), alternative splicing gives rise to isoforms such as CD33ΔE2, which lacks the V-set domain. This splicing difference has significant therapeutic implications because many current anti-CD33 therapies rely on binding to the V-set region; hence, as alternative isoforms appear, a need arises for modulators that can target either the common or alternative domains. The novel CD33PAN antibodies directed against the C2 domain described in recent studies are a good example of how structure–function insights can drive new molecule development.

CD33 in the Immune System

CD33 functions as an inhibitory receptor, playing a regulatory role by dampening the activation of immune cells. This receptor participates in modulating immune cell signaling pathways by recruiting phosphatases through its ITIMs. When CD33 is engaged by its ligands (typically presenting sialic acid tags on cell surfaces), it can inhibit cellular activities such as phagocytosis and cytokine secretion. In the context of neurodegeneration, particularly Alzheimer’s disease, genome‐wide association studies have associated specific variants of CD33 with altered microglial function and disease risk. Moreover, in hematological malignancies such as AML, CD33 is expressed on leukemic blasts and has been widely exploited as a therapeutic target due to its cell-specific expression and rapid endocytic behavior, which facilitates the internalization of antibody conjugates. Overall, CD33 plays multifaceted roles across various immune cell populations, and its inhibition or modulation has emerged as a powerful strategy to either boost immune clearance or tune destructive inflammation.

Recent Advances in CD33 Modulators

In recent years, significant progress has been made in the development of CD33 modulators. Researchers have expanded beyond traditional antibody–drug conjugates (ADCs) such as gemtuzumab ozogamicin to explore new molecular formats, including engineered antibodies, bispecific constructs, and small molecules that modulate splicing. These new molecules have been designed to overcome limitations inherent to targeting the V-set domain exclusively and to engage alternative epitopes as well as to provide improved potency and safety profiles.

Newly Identified Molecules

Multiple new molecules have been identified as promising CD33 modulators through a combination of molecular engineering, structure‐based design, and innovative screening strategies. One prominent example is the development of antibodies that target the membrane-proximal C2-set domain of CD33, dubbed “CD33PAN antibodies”. Unlike conventional antibodies that bind exclusively to the V-set domain, these novel antibodies can bind regardless of whether the V-set domain is present in its canonical form. This flexibility is particularly important given the existence of alternatively spliced isoforms (such as CD33ΔE2) that might not be accessible to earlier antibody therapies.

In addition to these domain-specific antibodies, various Fc-engineered anti-CD33 antibodies have been introduced. For instance, BI 836858, an Fc-engineered monoclonal antibody, has been studied for its ability to induce antibody-dependent cellular cytotoxicity (ADCC) and block CD33-mediated immune-suppressive signaling in myeloid-derived suppressor cells (MDSCs). Such molecules are designed to increase the functional response by modifying the Fc region (often through substitutions like SDIE) to enhance interactions with Fc receptors on natural killer (NK) cells and macrophages.

Another innovative strategy involves the generation of bispecific antibodies or bi-specific T cell engagers (BiTEs) that incorporate a CD33-binding arm linked to a CD3-binding moiety. These molecules, such as the one described in modular targeting systems and further characterized by the ability of CD33/CD3 bispecific antibodies, recruit T cells to engage and eliminate CD33+ tumor cells. They are particularly notable in the treatment of AML due to the need to efficiently target leukemic blasts while mitigating toxicity.

Cutting-edge research has also led to the discovery of small-molecule modulators that influence CD33 pre-mRNA splicing. These modulators are designed to shift the splicing profile of the CD33 gene, favoring the production of isoforms that are less inhibitory. By altering the RNA splicing machinery, these modulators represent a novel approach that could complement antibody-based strategies by impacting the quantity and quality of CD33 isoforms expressed on the cell surface.

Furthermore, several novel antibody conjugates and chimeric antigen receptor (CAR) constructs have emerged from the patent literature. These include chimeric molecules engineered to bind CD33 with enhanced internalization and improved pharmacokinetic properties. For example, engineered CD33-targeted immunotherapies incorporate modifications that allow for rapid endocytosis and intracellular payload delivery, thereby increasing cytotoxicity in cancer cells while potentially reducing off-target effects.

Finally, the identification of CD33 molecules that modulate the inhibitory signaling of the receptor has also been explored through surrogate systems such as reporter cell assays. These assays test the signaling outcome after antibody binding, such as induction of SYK phosphorylation, which helps in screening new molecules based on their capacity to internalize CD33 and inhibit its suppressive signals in a cell-based model.

Mechanisms of Action

The newly identified molecules for CD33 modulation exert their effects through several distinct mechanisms. First, the antibodies targeting the C2-set domain work by engaging regions of CD33 that are less affected by splicing variations. This allows them to bind to both full-length and splice variant forms of CD33, thereby overcoming one of the limitations seen in traditional therapies that target solely the V-set domain. These antibodies have been shown to internalize upon binding, which can lead to receptor degradation and downstream attenuation of inhibitory signaling. By removing the receptor from the cell surface, these antibodies can alleviate the suppression of phagocytosis and increase the clearance of pathogenic ligands or tumor cells.

Fc-engineered antibodies, such as BI 836858, are modified in their constant regions to enhance interactions with effector immune cells. This results in increased ADCC activity, meaning that natural killer cells and other Fc receptor-expressing immune cells more effectively recognize and kill target cells that express CD33. The engineering often involves substitutions like S239D/I332E that have been shown to improve binding to CD16 (FcγRIIIa). The enhanced ADCC effect effectively converts the CD33-expressing cells—from suppressive myeloid-derived cells to targets for immune-mediated cytotoxicity.

Bispecific antibodies take advantage of the modular design by bridging CD33 on tumor or myeloid cells with CD3 on T cells. This proximity induces T-cell receptor engagement and the formation of an immunological synapse, leading to T-cell activation and targeted cell lysis. Such constructs overcome limitations associated with low target antigen density by providing costimulatory signals through the inclusion of domains like CD137 ligand. These bispecific molecules thus exert a dual mechanism: they not only redirect T cells but also deliver secondary activation signals that enhance cytotoxicity under low antigen conditions.

Small-molecule splicing modulators affect CD33 expression at the RNA level. By binding to splicing regulatory elements, these modulators change the relative abundance of CD33 isoforms. Since the full-length CD33 isoform contains the V-set domain that is essential for its inhibitory function, altering the splicing pattern to favor isoforms lacking this domain (such as CD33ΔE2) can potentially reduce the net inhibitory signaling of CD33 in microglia and myeloid cells. Thus, these small molecules have the potential to indirectly reprogram the immune response by modulating the expression of functional CD33 on the cell surface.

Collectively, these mechanisms demonstrate a transition from simply using cytotoxic payloads (as in traditional ADCs) to engaging with the immune system through multiple modalities. The new molecules not only enhance internalization and degradation of CD33 but also actively engage cell-mediated immune responses to kill target cells, thereby broadening the therapeutic window across various disease indications.

Therapeutic Applications of CD33 Modulators

The therapeutic applications of novel CD33 modulators span several clinical areas, from oncology—particularly AML—to neurodegenerative conditions such as Alzheimer’s disease. There is growing evidence that properly modulated CD33 signaling can ameliorate disease progression by either lifting the brakes on phagocytosis in microglia or by directly targeting malignant myeloid cells in hematologic cancers.

Clinical Trials and Studies

A number of clinical studies and early-phase trials have investigated new CD33 modulators. For example, various Fc-engineered antibodies are being trialed to assess their safety and efficacy in reducing the suppressive function of MDSCs in myelodysplastic syndromes (MDS) and AML. Such studies take into consideration receptor density, internalization kinetics, and ADCC induction, which are critical parameters for clinical success. Additionally, bispecific antibodies that incorporate a CD33-targeting domain have been explored in clinical settings with early-phase trials focusing on their ability to recruit T cells, as reported in studies involving modular targeting systems and human AML cell line evaluations.

Moreover, several patents detail novel molecular formulations that are currently in various stages of preclinical and clinical development. These patents underscore the emphasis on next-generation CD33 modulators that can be utilized for either direct cancer therapy or as an adjunct to other immune-based therapies such as CAR-T cell treatments. Recent clinical experiences with gemtuzumab ozogamicin provided proof of concept that targeting CD33 could yield improved patient responses in AML. However, the newer molecules seek to enhance the therapeutic index by reducing on-target off-leukemia toxicity and by preserving normal myelopoiesis, as evidenced by studies on CD33-deleted hematopoietic stem cells that protect against myelosuppression.

In parallel, efforts have also been directed at harnessing CD33 modulation to impact the risk of neurodegenerative disorders. The protective association of certain CD33 genetic variants with Alzheimer’s disease has sparked interest in using modulators—both antibodies and small molecules—to shift CD33 expression profiles in microglia. These compounds may ultimately serve to increase phagocytosis of amyloid-beta peptides, thereby reducing plaque burden in the brain. Reporter cell assays that validate the functional readout of CD33 modulation—such as measuring SYK phosphorylation and intracellular calcium signaling in response to anti-CD33 antibodies—are critical in drawing the link between molecular action and anticipated in vivo benefits. Thus, the clinical studies exploring these endpoints are paving the way for a novel class of therapeutics aimed at modulating CD33 signaling in different disease contexts.

Potential Benefits and Risks

The potential benefits of these new CD33 modulators are multifold. In cancer therapy, especially in AML and MDS, novel CD33 targeting molecules promise higher selectivity and potency. By targeting alternative epitopes (for example, the membrane-proximal C2-set domain) rather than the conventional V-set region, new molecules exhibit broader reactivity against diverse CD33 isoforms produced by alternative splicing. This is anticipated to translate into improved internalization rates and better clearance of malignant cells while minimizing toxicity to normal hematopoietic progenitors.

Moreover, the use of Fc-engineered antibodies and bispecific constructs enables the harnessing of the patient’s own immune system—either via enhanced ADCC or by recruiting cytotoxic T lymphocytes—which is especially useful in cases where antibody monotherapy has limited efficacy. The incorporation of costimulatory signals in bispecific constructs further improves the therapeutic window, particularly in low antigen-expressing tumors. In the context of Alzheimer’s, shifting CD33 isoform expression through small-molecule splicing modulators may reduce the inhibitory signaling in microglia, enhancing amyloid-beta clearance and thus potentially altering disease progression.

On the risk side, while the modulation of CD33 holds great promise, it is not without challenges. The heterogeneous expression of CD33 across various cell types raises concerns about off-target effects. Although many novel modulators are designed to selectively target CD33 in malignant cells or overactivated microglia, unintended suppression or activation of normal myeloid functions could lead to adverse events such as excessive inflammation or compromised host defense. Moreover, in the clinical trials of CD33-targeting therapeutics, issues such as myelosuppression and off-target cytotoxicity have historically been observed. New molecules that enhance internalization may mitigate some of these risks, but they also necessitate careful dosing and monitoring strategies.

Another concern is the variability in CD33 isoform expression among patients, which can affect the efficacy of modulators that target specific domains. For instance, therapies based exclusively on V-set epitope recognition may not be effective in patients who predominantly express the CD33ΔE2 isoform, making the development of CD33PAN antibodies and small-molecule splicing modulators particularly attractive. The immune‐modulating therapies that rely on bispecific T cell engagement also require a careful calibration of effector‐to‐target ratios, and their success might depend on the patient’s T-cell repertoire and overall immune competence.

Challenges and Future Directions

The revolution in CD33 modulator design comes with evolving research challenges and promising future opportunities. Although new molecules have been identified and early clinical data are encouraging, multiple hurdles remain in optimizing these agents for safe and effective use in patients with diverse disease states.

Current Research Challenges

One of the primary challenges in the field is the heterogeneity of CD33 expression and isoform distribution. Since CD33 can exist as full-length or splice variants, and because the conventional therapeutic antibodies mainly recognize the V-set domain, there has been a pressing need for modulators that address this complexity. Novel molecules such as the CD33PAN antibodies address this issue by targeting the C2-set domain, but the translation of these preclinical findings into broadly effective clinical reagents requires further validation in larger and more diverse patient populations.

Another challenge is the species-specific differences in CD33 structure and signaling. For example, mouse CD33 exhibits differences not only in the extracellular region but also in the cytoplasmic domain compared to its human counterpart. These differences complicate the preclinical assessment of human-specific modulators in animal models, thereby necessitating the development of surrogate systems or humanized models to accurately predict clinical outcomes. As a result, the validation of new CD33 modulators demands sophisticated in vitro reporter cell assays and xenograft models that faithfully recapitulate human CD33 biology.

From a pharmacokinetic and pharmacodynamic perspective, the optimization of antibody engineering (including enhanced Fc-receptor binding) is critical to balance efficacy and safety. Although modifications such as the SDIE substitution have shown potential improvements in ADCC activity, determining the appropriate dosing, infusion schedules, and managing immune complex formation remain areas of active research. Variations in circulating CD33 levels and receptor occupancy also complicate the design of effective dosing regimens, urging researchers to employ more robust pharmacokinetic-pharmacodynamic modeling and to refine dosing strategies based on target modulation readouts.

The design and clinical testing of small-molecule modulators that alter CD33 pre-mRNA splicing is an emerging area with significant challenges. The specificity of these small molecules for their intended splicing regulatory elements, off-target effects on alternative splicing pathways, and the long-term consequences of altering CD33 isoform ratios in various cell types require significant investigative efforts. As such, the development of these molecules necessitates a comprehensive understanding of the splicing machinery along with advanced high-throughput screening and validation assays.

Additionally, the combination of CD33 modulators with other therapies, such as chemotherapy, immune checkpoint inhibitors, or CAR-T cell treatments, poses logistical and biological challenges in terms of synergy and toxicity management. The immunomodulatory nature of these agents means that even slight variations in timing, dosing, or patient immune status could have profound effects on treatment outcomes. Therefore, rigorous clinical trials with robust biomarker analyses are essential to optimize combination regimens and to mitigate the risks associated with unanticipated immune side effects.

Future Research Opportunities

Looking forward, there are myriad opportunities for further advances in the field of CD33 modulation. The integration of high-resolution structural biology with computational modeling is poised to drive the rational design of even more potent modulators. Future research could focus on obtaining atomic resolution structures of CD33, particularly its transmembrane and intracellular domains, to better understand how novel antibodies, Fc-engineered molecules, or small molecules interact with the receptor. Such detailed structural insights could facilitate the design of next-generation modulators that are more selective, have rapid internalization kinetics, and exhibit improved safety profiles.

The development of more physiologically relevant model systems—such as humanized mouse models or advanced in vitro systems using patient-derived cells—represents another promising research avenue. These models will enable researchers to test new molecules in environments that closely mimic human disease states and allow for better predictions of clinical efficacy. In Alzheimer’s research, for instance, the use of patient-derived microglia or induced pluripotent stem cell (iPSC)-derived models could help elucidate how CD33 splicing modulators affect amyloid-beta clearance and microglial activation.

Furthermore, comprehensive molecular studies examining the interplay between CD33 and other immune receptors (such as TREM2) could uncover potential synergistic targets. Evidence suggests that inhibitory signaling through CD33 may counteract the activating functions of receptors like TREM2, and modulating this balance might enhance the overall immune response against pathological targets in both cancer and neurodegenerative diseases. Future studies could leverage systems biology approaches and single-cell transcriptomics to refine our understanding of these interactions and to identify additional modulatory nodes in the network.

On the clinical front, the continued evaluation of bispecific antibodies represents an area with tremendous potential. With early clinical data indicating that bispecific CD33/CD3 molecules can effectively redirect T cells to engage and kill target cells, there remains the opportunity to optimize these molecules further. Advancements in antibody engineering, including the incorporation of costimulatory domains (for example, CD137 ligand) into the targeting modules, are likely to enhance T cell activation even in cases of low CD33 expression on target cells. Future clinical trials that stratify patients based on CD33 isoform expression and target density could provide key insights that drive personalized therapeutic strategies.

In the realm of small-molecule splicing modulators, future research can concentrate on high-throughput screens to identify lead compounds with optimal specificity and minimal off-target effects. Refining these molecules will require iterative medicinal chemistry and detailed pharmacodynamic studies that focus on safely altering CD33 isoform ratios in specific cell subsets. These efforts could eventually lead to dual-modality treatments where modulation of splicing is combined with immunotherapeutic mechanisms to restore normal phagocytic function in microglia, thereby altering the course of Alzheimer’s disease.

Emerging clinical trials incorporating these new modulators offer an important window of opportunity to integrate preclinical advances with patient care. The patent literature provides numerous examples of novel CD33-binding antibodies and CAR constructs that are in early-stage clinical development. Continued collaborative efforts between academic researchers, pharmaceutical companies, and clinical investigators will accelerate the translation of these molecules into approved therapies. Moreover, a better understanding of the mechanisms of action of these molecules will help in designing combination therapies that enhance their efficacy while minimizing adverse effects.

Finally, the future of CD33 modulation may benefit from novel delivery platforms. For example, nanoparticle-based systems or exosome-mediated delivery of antibody fragments or small-molecule splicing modulators could improve tissue penetration and target engagement in both hematological malignancies and neurodegenerative diseases. These advanced delivery systems hold the potential to eliminate some of the pharmacokinetic hurdles currently limiting the efficacy of large biomolecules in clinical practice.

In summary, future research opportunities in CD33 modulation are abundant and include structural elucidation, improved preclinical modeling, combination therapy optimization, and the development of innovative delivery systems. All these directions converge toward the goal of refining the molecular toolkit available for modulating CD33 function in a manner that is both safe and clinically effective.

Conclusion

In a general sense, the landscape of CD33 modulators has evolved significantly in recent years thanks to advances in molecular targeting and antibody engineering. From our broad research on the structure and function of CD33, new molecules have been designed that target alternative epitopes beyond the traditional V-set domain. Specifically, the development of CD33PAN antibodies targeting the membrane-proximal C2 domain, Fc-engineered antibodies such as BI 836858 that enhance ADCC, and innovative bispecific antibodies bridging CD33 with CD3 all represent breakthroughs that illustrate a balanced approach to modulating both immune suppression in cancers and defective microglial activity in neurodegeneration. Meanwhile, small molecule modulators that influence CD33 pre-mRNA splicing emerge as a unique strategy to shift isoform expression towards a more therapeutically favorable profile.

On a more specific level, these new molecules offer mechanisms of action that include receptor internalization and degradation, augmented effector cell recruitment, and targeted modulation of gene splicing—all designed to improve clinical outcomes by overcoming limitations seen in earlier therapies such as gemtuzumab ozogamicin. Clinical trials and early-phase studies provide promising evidence that these novel agents can safely and effectively modify disease progression in both hematological malignancies like AML and in neurodegenerative conditions such as Alzheimer’s disease.

From a general perspective, while the benefits of these new CD33 modulators are multifaceted, there remain significant challenges. Variability in CD33 isoform expression, species-specific differences in receptor structure, and the intricate balance between immune activation and suppression all necessitate further research. Future opportunities lie in refining the structural insights of CD33, optimizing preclinical models, and designing combination therapies that integrate these modulators with other agents to maximize therapeutic efficacy.

In conclusion, the new molecules for CD33 modulation—including novel antibody constructs (CD33PAN, Fc-engineered variants, and bispecific antibodies), small-molecule splicing modulators, and innovative chimeric constructs—represent a sophisticated and promising portfolio aimed at rebalancing immune inhibition and activation. Their development is informed by detailed molecular insights and is driven by the clinical need for more effective and selective therapies. The detailed mechanistic understanding, combined with advances in delivery systems and clinical trial strategies, holds the promise of significantly altering the treatment landscape for diseases where CD33 plays a pathogenic role. This multi-perspective approach—from the molecular structure and function to therapeutic applications and future research directions—ensures that the field will continue to innovate, address existing challenges, and ultimately provide safer and more effective treatments for patients.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成