What are the therapeutic applications for HER3 modulators?

11 March 2025
Introduction to HER3
HER3, also known as ErbB3, is a member of the human epidermal growth factor receptor (HER/ErbB) family that plays a central role in cell signaling. It is unique among the family members due to its catalytically impaired kinase domain, yet it can potently activate downstream pathways when paired with other receptors. The study of HER3 has grown exponentially over the past several years, as researchers have elucidated its role in both normal physiology and disease pathology. In this section, we provide a broad overview of its structure and function before discussing its pathological relevance.

HER3 Structure and Function
The HER3 receptor comprises a large extracellular domain with several distinct subdomains that facilitate ligand binding. Despite its lack of intrinsic kinase activity—a characteristic that differentiates it from other members such as HER1 or HER2—HER3 has multiple phosphorylation sites on its cytoplasmic tail. These sites become phosphorylated upon heterodimerization with active partners (most notably HER2), thereby recruiting key signaling molecules like the p85 regulatory subunit of phosphatidylinositol 3‑kinase (PI3K). This unconventional mode of activation is crucial because it allows HER3 to effectively integrate signals essential for cell survival, proliferation, and differentiation. Structural studies using crystallography and advanced molecular modeling have provided insight into its extracellular binding conformations and cytoplasmic interactions, making HER3 a complex yet highly targetable receptor.

Role of HER3 in Disease Pathology
Although under normal conditions HER3 plays roles in tissue development and cellular differentiation, its aberrant expression or activation is implicated in several disease states. In many cancers, HER3 expression is either upregulated or becomes hyperactivated through the formation of heterodimers with HER2 or EGFR. This process is closely associated with resistance to therapies that target other members of the HER family and mediates activation of the potent PI3K/AKT pathway, a critical signaling cascade involved in cell survival and proliferation. In addition, HER3’s involvement extends to tumor progression and metastasis, rendering it a compelling target for drug development. Apart from oncology, emerging evidence suggests that HER3’s dysregulation might also play a role in certain non-malignant conditions, although these applications are less extensively studied compared to cancer.

HER3 Modulators
HER3 modulators encompass a range of therapeutic agents designed to directly modulate the receptor’s function or its downstream signaling. Given HER3’s role as a critical dimerization partner and mediator of resistance, researchers have developed numerous molecular modalities to inhibit its function. These modulators are engineered to block ligand binding, prevent dimerization, or deliver cytotoxic payloads selectively to HER3-expressing cells.

Types of HER3 Modulators
The therapeutic agents targeting HER3 can be classified into several categories:

1.  Antibody‐drug conjugates (ADCs): These are complex molecules that combine an anti‑HER3 antibody with a cytotoxic drug payload. Examples include candidates such as SHR‑A2009, JSKN016, IBI‑133, DB‑1310, and SIBP‑A13. ADCs are engineered to recognize HER3 on the surface of tumor cells and deliver cytotoxic agents directly, thereby minimizing off‑target effects. Some ADCs are designed to inhibit both HER3 and additional targets (e.g., HER3 x Trop‑2 or HER3 x TOP1) to overcome resistance mechanisms.

2.  Engineered antibody fragments and affibody molecules: These small proteins offer advantages of rapid tissue penetration and fast clearance from non‑target areas. They can be multimerized (engineered for multivalency) to induce receptor downregulation or internalization, which further limits the HER3 signaling. Studies have shown that multivalent affibodies can more efficiently inhibit heregulin‑induced signaling, leading to enhanced anti-proliferative activities in cancer cell lines.

3.  Monoclonal antibodies (mAbs): Numerous anti‑HER3 mAbs have been designed to block ligand binding and receptor activation. Examples include those described in patents, which emphasize antibodies that target the HER3 extracellular domain with mechanisms that prevent activation even in the presence of its ligand, heregulin. Such antibodies often work by sterically hindering the receptor dimerization, or by accelerating receptor internalization and degradation.

4.  Small molecule inhibitors: Although HER3 lacks intrinsic kinase activity, small molecule inhibitors that indirectly modulate HER3 activity by interfering with receptor–partner interactions or the stabilizing chaperones are also under exploration. These molecules can be designed to disturb the scaffolding interactions or to mimic inhibitory peptides that disrupt HER3 complex formation.

Mechanisms of Action
HER3 modulators act through multiple interrelated mechanisms, and their therapeutic function depends on their molecular design and mode of delivery:

1.  Ligand Blocking: Many HER3 modulators function by competitively inhibiting the binding of ligands such as heregulin. By occupying the HER3 extracellular domain, these modulators prevent its activation and subsequent dimerization with other receptors. This leads to effective attenuation of downstream oncogenic signaling pathways.

2.  Receptor Dimerization Inhibition: Some engineered antibodies or bispecific molecules bind simultaneously to HER3 and its dimerization partners (e.g., HER2), “locking” the receptor into inactive dimers. This sequestration not only blocks activation but also prevents the formation of signaling complexes, thereby hindering the PI3K/AKT pathway.

3.  Induced Receptor Internalization and Degradation: Certain modulators, particularly multivalent constructs and ADCs, are designed to promote rapid internalization of HER3. Once internalized, the receptor is directed to lysosomal degradation, causing downregulation of HER3 on the cell surface and prolonged suppression of its signaling activity.

4.  Cytotoxic Payload Delivery: ADCs attach a potent cytotoxic agent to the HER3-targeting antibody. After binding to HER3, the ADC is internalized, and the payload is released intracellularly, leading to cell death. This dual mechanism not only blocks HER3 signaling but also directly kills tumor cells that overexpress HER3.

5.  Allosteric Modulation: Emerging strategies include agents that induce conformational changes in HER3, rendering its intracellular tail less accessible for phosphorylation. Such allosteric modulators may offer improved efficacy by subtly altering the receptor configuration, though these approaches are still in preclinical evaluation.

Therapeutic Applications of HER3 Modulators
HER3 modulation has been predominantly explored in oncology due to the strong association between HER3 signaling and tumor progression. However, potential applications in neurological disorders and other disease areas are under investigation as well.

Oncology
In oncology, HER3 modulators have emerged as promising therapeutic agents due to their significant roles in tumor survival, resistance to current therapies, and metastatic progression.

•  Targeting resistance mechanisms: HER3 is often upregulated as an adaptive response to therapies targeting HER1/EGFR and HER2. Its increase in signaling activity is one of the primary culprits in acquired drug resistance, for example in breast cancer and lung cancer. By inhibiting HER3 directly, modulators can overcome resistance and enhance the effectiveness of combination therapies.
•  Combination therapies: HER3 modulators are frequently used in combination with other targeted agents. ADCs that incorporate HER3-targeting antibodies may be coupled with chemotherapeutic drugs, while bispecific antibodies that engage both HER2 and HER3 can result in a synergistic suppression of cell survival signals. This approach is particularly valuable in HER2-positive cancers where HER3 forms heterodimers.
•  Broad spectrum of cancers: Preclinical studies have shown that HER3 modulators can be effective in several tumor types. These include:

  – Breast Cancer: HER3 overexpression and activation are frequently observed in breast cancer, especially in HER2-positive and hormone receptor-positive subtypes. Research indicates that targeting HER3 can disrupt critical survival pathways, enhance the efficacy of anti-HER2 agents like trastuzumab and pertuzumab, and overcome endocrine resistance in ER-positive tumors.
  – Lung Cancer: In non-small cell lung cancer (NSCLC), HER3 is implicated in tumor growth and in mediating resistance to EGFR inhibitors. Modulators that block HER3 can improve outcomes and potentially serve as biomarkers for patient selection.
  – Colorectal Cancer: HER3’s role in dimerization with other HER receptors makes it a potential target in colorectal cancer. Early-phase clinical assessments with HER3-targeting mAbs and ADCs suggest that patients with high HER3 expression may benefit significantly from these agents.
  – Pancreatic Cancer: Several ADCs targeting HER3 have been evaluated in pancreatic cancer xenograft models, demonstrating significant tumor regression and improved survival in preclinical studies.
  – Other solid tumors: Research continues into the applicability of HER3 modulators in ovarian cancer, head and neck cancers, and certain lung tumors. Their potential to block HER3 signaling makes them attractive candidates irrespective of tumor origin, especially when used in combination with other targeted therapies.

•  Biomarker-driven strategies: By measuring levels of HER3 expression, phosphorylation status, or the formation of heterodimers (e.g., HER2-HER3 complexes), clinicians can stratify patients to identify those most likely to benefit from HER3 modulators. Such biomarker-guided approaches have been validated in preclinical imaging studies employing radiolabeled affibodies and antibody fragments for PET imaging.
•  Resistance reversal: Beyond primary anticancer activity, HER3 modulators serve as tools to reverse therapeutic resistance. For instance, when tumor cells escape therapy by upregulating HER3, the addition of HER3-targeted agents can restore sensitivity to other inhibitors, presenting an integrated treatment option.

Neurological Disorders
While the oncology applications of HER3 modulators are the most advanced, emerging research suggests possible applications in neurological disorders, although this area is less developed. The rationale for potential application in the central nervous system (CNS) is based on several perspectives:

•  Neuroprotective strategies: In normal neural tissue, HER3 and its partners in the HER receptor family contribute to neural development and survival signaling. Dysregulation of these pathways may underlie aspects of neurodegenerative disorders. Modulating HER3 might be harnessed to protect neurons against apoptosis or dysfunction, thus offering a novel neuroprotective strategy.
•  Altering inflammatory responses: Chronic neural inflammation is a contributing factor in many neurodegenerative diseases. Certain HER3 modulators, by modulating associated downstream pathways (e.g., PI3K/AKT), might help balance neuroinflammatory processes or protect neural networks from injury.
•  Indirect effects on cognitive function: Although not yet clinically validated, early research into HER3-targeted therapies has raised the possibility that manipulating HER3 signaling could influence synaptic plasticity and neuronal survival, which are essential factors in disorders such as Alzheimer’s disease or other cognitive deficits.

At this time, neurological applications remain largely theoretical and require more substantial preclinical exploration before any translation into clinical trials for CNS indications.

Other Potential Applications
Beyond oncology and neurology, there are speculative domains where HER3 modulators could be applied:

•  Inflammatory disorders: Given the cross-talk between HER signaling and inflammatory cascades, modulating HER3 might have ripple effects in reducing pro-inflammatory cytokine expression. This could be in settings such as autoimmune disorders or chronic inflammatory diseases where aberrant cell survival signals contribute to pathology.
•  Metabolic regulation: Some studies have hinted at relationships between HER receptor activity and metabolic processes, although HER3’s direct involvement is less clear. There is potential for future research to decipher links between HER3 modulators and conditions like type 2 diabetes or metabolic syndrome, particularly if HER3 is found to influence insulin receptor signaling or energy metabolism indirectly.
•  Developmental disorders: Since HER3 is implicated in cellular differentiation and organ development, there may be unexplored roles for HER3 modulators in treating congenital disorders that involve aberrant HER3 signaling. As our understanding of HER3’s developmental roles increases, there might be niche applications for modulators in regenerative medicine or tissue repair strategies.

Clinical Trials and Research
The journey of HER3 modulators from bench to bedside has involved extensive preclinical studies followed by a growing number of clinical trials designed to measure safety, efficacy, and pharmacokinetic profiles. Research efforts highlight that HER3 modulators are not only useful as monotherapies but are also highly effective when combined with other targeted agents or chemotherapeutic drugs.

Current Clinical Trials
Clinical trials investigating HER3-targeted agents have employed various trial designs and patient populations. For instance, several ADCs and antibody fragments have entered Phase 1/2 and even Phase 3 trials to assess their efficacy in tumors with high HER3 expression. Examples include:

•  HER3-targeting ADCs being evaluated in both neoadjuvant and advanced settings in breast cancer, as well as in other solid tumors. One window-of-opportunity study focusing on U3‑1402—a HER3-targeted ADC—aims to correlate response with ERBB3 (HER3) expression levels in operable breast cancer patients.
•  Early-phase studies employing radiolabeled affibodies and antibody fragments for PET imaging have been implemented to measure HER3 expression and therapeutic target engagement in real time. This strategy helps refine patient selection and dosing strategies.
•  Combination trials wherein HER3 modulators are used alongside established anti-HER2 therapies or PI3K inhibitors. The rationale for such combinations stems from resistance mechanisms in cancers that simultaneously upregulate multiple HER family receptors.

These studies are meticulously designed with biomarker-driven endpoints, integrating molecular imaging and tissue analysis to validate HER3 downregulation, receptor occupancy, and downstream pathway inhibition.

Results and Findings
Preliminary results from clinical studies involving HER3 modulators are promising, particularly in oncology, where they have demonstrated:

•  Tumor growth inhibition and even regression in xenograft models, showing better efficacy compared with standard treatments when HER3 modulators are added.
•  Pharmacodynamic evidence that effective HER3 modulation correlates with a decrease in phosphorylated HER3, reduced AKT signaling, and increased receptor internalization and degradation.
•  Enhanced tumor imaging contrast, which helps in real-time assessment of HER3 receptor status during treatment. These imaging studies, typically carried out using PET or SPECT with radiolabeled agents, confirm the specific binding and targeting of HER3 by these modulators.
•  Favorable safety profiles in Phase I studies; in some cases, systemic exposure was greater than dose-proportional, indicating a target-mediated drug disposition. However, dose escalation studies have been critical to determine the therapeutic window while minimizing off-target effects.

Overall, the data indicate that HER3 modulators can effectively reduce tumor cell viability, overcome resistance mechanisms, and, when used in combination, may offer synergistic benefits. These findings are reinforced by a number of preclinical and clinical studies, providing substantial evidence for their potential in personalized therapeutic strategies.

Challenges and Future Perspectives
Despite the promising therapeutic applications of HER3 modulators, a number of challenges remain. Continued research and refinement of these agents will likely pave the way for improved clinical outcomes.

Current Challenges in HER3 Modulator Development
Several issues need to be resolved in order to fully harness the therapeutic potential of HER3 modulators:

•  Biomarker Identification: One of the critical challenges is the lack of universally accepted biomarkers to stratify patients. Although HER3 mRNA levels, receptor phosphorylation status, and the presence of HER2-HER3 heterodimers have been studied, there is still a need for robust, predictive biomarkers. This will help in selecting the right patient populations and predicting therapeutic response.
•  Drug Resistance: As HER3 is often implicated in resistance mechanisms against EGFR and HER2 inhibitors, further studies are necessary to understand how tumors evolve resistance to HER3-targeted therapies. The influence of compensatory signaling through alternative pathways remains a critical area of investigation.
•  Pharmacokinetics and Bio-distribution: Due to HER3’s expression in both tumors and normal tissues, ensuring that modulation is specific to cancerous tissues without harming normal cells poses a significant challenge. This is particularly important for ADCs and multivalent molecules, which must be carefully balanced to provide adequate tumor uptake while minimizing systemic toxicity.
•  Optimization of Drug Formats: While ADCs, mAbs, and small molecules have each shown promise, their optimal design—including binding affinity, valency, and conjugation methods—remains an active area of research. The differences in receptor internalization rates and biodistribution must be carefully addressed to enhance clinical efficacy.
•  CNS Penetration for Non-oncologic Indications: For the potential application in neurological disorders, achieving adequate blood-brain barrier penetration and minimizing off-target effects in the CNS pose significant technical challenges.
•  Safety and Tolerability: Although early-phase trials have demonstrated acceptable safety profiles, long-term safety data are limited. The potential for immunogenic reactions, off-target effects, or unexpected toxicities requires ongoing vigilance during clinical development.

Future Research Directions
Looking ahead, several key areas of research are likely to guide the development and application of HER3 modulators:

•  Enhanced Biomarker Discovery: Future clinical trials will need to incorporate genomic, transcriptomic, and proteomic data to refine biomarker identification. This will enable more precise patient stratification and real-time monitoring of HER3 expression and activity during treatment.
•  Combination Therapy Development: Given the complexity of HER signaling networks, combination therapies that target multiple HER family members (e.g., HER2/HER3 combinations) or integrate inhibitors of downstream pathways (like PI3K/AKT inhibitors) will become increasingly important. These approaches are especially promising for overcoming resistance and prolonging patient survival.
•  Advanced Drug Engineering: The future will likely see improved designs of HER3 modulators that combine the benefits of different molecular formats. For example, engineering bispecific antibodies that not only block HER3 but simultaneously engage immune effector mechanisms or enhance payload delivery could result in increased anti-tumor activity.
•  Novel Delivery Platforms: Emerging technologies such as nanoparticle-based drug delivery systems or engineered viral vectors might be harnessed to deliver HER3 modulators more efficiently to tumors, thereby improving pharmacokinetics and reducing systemic toxicity.
•  Exploration of Non-oncology Applications: Although the primary focus remains oncology, deeper mechanistic studies on HER3’s role in the nervous system may open up new opportunities in neuroprotection and neuroinflammatory regulation. This will require collaborating across disciplines to adapt modulation strategies for CNS penetration and tissue specificity.
•  Personalization of Therapy: As precision medicine advances, tailoring HER3 modulators to patients based on the genetic and molecular profiles of their tumors will be critical. Integrating patient-derived xenograft models and organoid cultures in preclinical testing could accelerate this personalized approach.
•  Long-term Safety and Resistance Studies: Additional research is imperative to understand long-term safety, especially in chronic treatments, and to monitor the potential emergence of secondary resistance mechanisms. Such studies will inform dose optimization, treatment duration, and the design of sequential therapies.

Conclusion
In summary, HER3 modulators represent a versatile class of therapeutics with significant implications for oncology, where they are already showing promise in overcoming resistance, enhancing targeted therapy efficacy, and providing options for patients with a wide range of solid tumors. The unique structural attributes of HER3—its impaired intrinsic kinase activity combined with its potent activation of downstream signaling when dimerized—have made it an attractive target for innovative drug design. Agents such as ADCs, monoclonal antibodies, engineered antibody fragments, and small molecule inhibitors each offer distinct mechanisms of action: from blocking ligand binding to inducing receptor internalization and delivering cytotoxic payloads.

From an oncological perspective, these modulators are making headway in the treatment of breast, lung, colorectal, and pancreatic cancers, among others, by targeting HER3-driven survival and proliferation pathways. Their application is further enhanced when combined with other targeted therapies such as HER2 inhibitors or PI3K/AKT inhibitors, as resistance remains a major hurdle in current treatment regimens. Additionally, early-stage clinical trials and molecular imaging studies have provided encouraging results regarding safety, pharmacodynamic responses, and effective tumor targeting.

Outside of oncology, while research into neurological and other non-cancer applications of HER3 modulators is still in its infancy, there is theoretical potential for neuroprotective effects and modulation of inflammatory processes in the central nervous system. Other potential applications, including metabolic regulation and treatment of certain inflammatory or developmental disorders, remain promising areas for future research.

Nonetheless, challenges including biomarker identification, drug delivery, optimal design of drug formats, and long-term safety must be addressed. Future research directions call for integrated biomarker discoveries, advanced drug engineering, and personalized therapy approaches that can effectively translate preclinical successes into durable clinical benefits. In the coming years, multidisciplinary collaboration and the adoption of innovative delivery platforms are expected to further enhance the effectiveness and applicability of HER3 modulators, ultimately revolutionizing precision medicine in oncology and beyond.

In conclusion, HER3 modulators stand at the intersection of sophisticated molecular biology and clinical innovation. Their development not only underscores the complexity of cancer signaling but also opens new avenues for treating resistant and aggressive tumors, marking a pivotal step towards more personalized and effective therapeutic strategies. Advances in preclinical models, emerging clinical trial results, and continued refinements in molecular design all contribute to an optimistic outlook for the future of HER3-targeted treatments.

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.