What drugs are in development for Hormone receptor positive HER2 negative breast cancer?

12 March 2025
Overview of Hormone Receptor Positive HER2 Negative Breast CancerDefinitionon and Characteristics
Hormone receptor–positive, HER2–negative breast cancer is defined by the expression of estrogen receptor (ER) and/or progesterone receptor (PR) in the absence of overexpression or amplification of the HER2 proto‐oncogene in cancer cells. This subtype is characterized by its hormone dependence, where the proliferation and survival of the tumor are largely driven by estrogen signaling. These tumors often exhibit a relatively lower proliferation rate compared to HER2+ or triple-negative forms, and they tend to be more differentiated histologically. However, their clinical behavior is heterogeneous due to additional molecular alterations within cell cycle regulation, growth factor signaling, and epigenetic modifications.

Epidemiology and Impact
HR+ HER2– breast cancer is the most common subtype of breast cancer, comprising around 70% of all cases. Given its high prevalence, these cancers have a significant impact on public health, both in terms of mortality and quality of life. Although patients with HR+ HER2– tumors often have a better initial prognosis and slower disease progression than other subtypes, long-term outcomes are complicated by the eventual development of endocrine resistance. The high incidence coupled with the potential for late relapse has been a driving force for research into improved therapeutic—including targeted—options to further enhance patient survival and quality of life.

Current Treatment Landscape

Existing Therapies
The current standard of care for HR+ HER2– breast cancer primarily involves endocrine therapies that block estrogen production or directly interfere with estrogen receptor signaling. These treatments include:
• Aromatase inhibitors (e.g., letrozole, anastrozole, exemestane) that reduce circulating estrogen levels for postmenopausal women.
• Selective estrogen receptor modulators (SERMs) such as tamoxifen, which bind to ER in a tissue-specific manner to block the proliferative estrogen effect.
• Selective estrogen receptor degraders (SERDs) such as fulvestrant that not only block the receptor but also promote its degradation.

In addition, combination regimens—particularly with cyclin-dependent kinase 4/6 (CDK4/6) inhibitors (such as palbociclib, ribociclib, and abemaciclib)—have become the mainstay in the first-line treatment setting by significantly prolonging progression‐free survival when combined with endocrine therapy. These drugs have transformed management by directly interfering with cell cycle progression in hormone-driven cancer cells.

Limitations of Current Treatments
Despite the significant advances made with endocrine therapy and the use of CDK4/6 inhibitors, several challenges remain. The onset of resistance—either de novo or acquired—is one of the primary caveats in HR+ HER2– treatment. Over time, tumor cells may acquire mutations (for example, in the ESR1 gene) or activate alternate signaling pathways (such as the PI3K/AKT/mTOR pathway) that confer resistance to hormone blockade. This resistance limits the long‐term efficacy of existing treatment regimens and ultimately necessitates the development of novel agents that can overcome or circumvent these mechanisms. Additionally, while improvements in response rates have been achieved, many patients eventually progress on therapy, emphasizing a clear need for drugs that can either provide a new mechanism of action or potentiate the activity of endocrine drugs without overlapping toxicities.

Drugs in Development

For HR+ HER2– breast cancer, several new therapeutic agents are evolving from the basic science insight into resistance mechanisms. New drugs are being evaluated in clinical trials and preclinical studies, and they can generally be categorized into novel hormone therapies, targeted therapies, and emerging immunotherapeutics.

Novel Hormone Therapies
New endocrine agents or improved versions of existing hormone therapies are being designed to counter resistance mechanisms and offer more potent blockade of estrogen signaling. They include:
• Next-generation SERDs that are orally bioavailable. Traditional fulvestrant, while effective, is given intramuscularly and may have limited bioavailability; newer oral SERDs are designed to offer improved convenience and enhanced degradation of the ER even in the presence of ESR1 mutations.
• Agents that modulate the co-activators of estrogen receptor transcription. Some of these investigational compounds are designed to interfere with the molecular machinery that governs hormone receptor activity, such as inhibitors of chaperone molecules or selective modulators targeting receptor co-activators. These agents aim to disrupt estrogen signaling at multiple levels, not just receptor binding.
• Drugs that combine both endocrine and other pathway inhibition. For instance, compounds that have dual activities, acting as SERDs and simultaneously inhibiting downstream effectors like molecules in the PI3K/AKT/mTOR pathway, are under early clinical evaluation. The goal is to create agents that address several resistance mechanisms simultaneously.
These novel hormone therapies are especially important for patients who harbor ESR1 mutations or have poor responses to conventional therapy. Their development is driven by the need to obtain complete receptor degradation and overcome hormone resistance, as indicated by recent clinical trials and research findings.

Targeted Therapies
Targeted therapies in development for HR+ HER2– breast cancer have broadened from simply combining endocrine therapy with CDK4/6 inhibitors. New agents are being developed that target specific pathways implicated in endocrine resistance:
• PI3K/AKT/mTOR inhibitors are at the forefront of targeted drug development. Given the well‐documented role of PI3K pathway activation in endocrine resistance, inhibitors that selectively target PI3K isoforms or dual inhibitors that block both PI3K and mTOR are being evaluated to restore hormone sensitivity. For example, alpelisib—a PI3K inhibitor that specifically targets the p110α isoform—is already approved in combination with endocrine therapy in some settings, but next‐generation PI3K inhibitors with improved selectivity and reduced toxicity profiles are also in development.
• Novel CDK4/6 inhibitors continue to be advanced. Although palbociclib, ribociclib, and abemaciclib have been integrated into clinical practice, additional compounds with different pharmacokinetic and safety profiles are being developed. These agents are being tested in series of clinical trials to determine whether they might further improve outcome either as monotherapy or in combination with other targeted agents.
• Agents targeting the FGFR (fibroblast growth factor receptor) pathway. Overexpression or activation of FGFR signaling has been implicated in resistance to endocrine therapies in some subsets of HR+ patients. Several FGFR inhibitors are undergoing early‐phase clinical evaluation as well as combination studies with endocrine therapy, aiming to disrupt this bypass survival route.
• ER co-targeting molecules that inhibit receptor cross-talk. Some novel molecules aim to disrupt signaling crosstalk between the estrogen receptor and other growth factor receptors or cell cycle regulators. For example, investigational compounds that inhibit both the ER and cyclin-dependent kinase signaling concurrently have been reported to show promising preliminary data.
• Multiplexed targeting strategies. Combining targeted therapies that address multiple resistance nodes simultaneously is another strategy under development. Several ongoing clinical trials evaluate the combination of endocrine therapy with inhibitors of both the PI3K pathway and CDK4/6, offering the potential for synergistic effects and prolonged disease control.
These targeted therapies are built on a molecular understanding of resistance pathways in HR+ HER2– breast cancer, and many are progressing from preclinical studies into early-phase clinical trials with promising early efficacy and manageable toxicity profiles.

Immunotherapies
Historically, immunotherapy in HR+ HER2– breast cancer has been less emphasized compared with triple-negative and HER2-positive subtypes because HR+ tumors are generally considered less immunogenic. Nonetheless, there is a growing body of research suggesting that the immune microenvironment may play a role in endocrine resistance and disease progression in HR+ breast cancer. Drugs and strategies in development include:
• Immune checkpoint inhibitors. Investigational studies are underway to see whether checkpoint blockade (for example, PD-1 or PD-L1 inhibitors) might have a role when combined with endocrine agents or targeted therapies. Although their single-agent activity in HR+ breast cancer is modest, combination therapies may enhance anti-tumor immunity and overcome resistance.
• Therapeutic vaccines. There are approaches aiming to stimulate an active immune response against tumor-associated antigens found in hormone receptor–positive cancers. These vaccines are designed to boost T-cell–mediated recognition of cancer cells, potentially improving long-term disease control when combined with other therapies.
• Adoptive T-cell therapies. Although still largely in early experimental phases for HR+ breast cancer, some studies are exploring the feasibility of adoptive cell transfer methods that can target specific neoantigens present in endocrine-resistant tumors.
Immunotherapeutic strategies for HR+ HER2– breast cancer are still in a nascent stage compared to other subtypes, but evolving techniques to induce immunogenic modulation in these “cold” tumors raise the possibility that immunotherapies—alone or in combination with endocrine agents—could become part of the future therapeutic armamentarium.

Clinical Trials and Research

Ongoing Clinical Trials
Many of the agents described above are being tested in rigorous clinical trials. Current studies frequently use adaptive, biomarker-driven designs to implement personalized treatment approaches. For example:
• Several phase I/II trials are evaluating novel oral SERDs in both treatment-naïve and endocrine-resistant populations.
• New PI3K inhibitors and dual PI3K/mTOR inhibitors are being studied in combination with endocrine therapy in phase II studies designed to select patients with PI3K pathway mutations.
• Trials combining next-generation CDK4/6 inhibitors with either endocrine therapy or additional targeted agents are ongoing to improve progression-free survival rates.
• Early-phase trials investigating FGFR inhibitors in patients with documented FGFR pathway activation are also underway.
• Immunotherapy investigations are seeing trials that combine PD-1/PD-L1 blockade with endocrine or CDK4/6 inhibitors to determine if synergistic effects can be realized in this population.
These trials often incorporate rigorous biomarker assessments (e.g., ESR1 mutation status, PIK3CA mutations, FGFR alterations) which help in patient stratification and in understanding the molecular determinants of response.

Recent Research Findings
Recent published data and conference abstracts have provided detailed insights into these drugs under development:
• Preclinical models have demonstrated that novel oral SERDs overcome ESR1 mutation–mediated resistance by achieving complete receptor degradation, thereby improving endocrine response.
• Studies have also reported that the combination of endocrine therapy with PI3K inhibitors leads to marked re-sensitization of tumors that had previously become refractory to hormone therapy, with significant improvements in progression-free survival in early-phase trials.
• Research on FGFR inhibitors has indicated potential benefit in a subset of patients with FGFR alterations, supporting the concept of tailoring therapy based on genomic profiling.
• Data emerging from early immunotherapy combination trials show hints of an immune activation signature even in hormone receptor–positive breast cancers, suggesting that these tumors may not be as “cold” as previously thought and that immune checkpoint inhibition in combination with targeted therapies may eventually enhance outcomes.
Moreover, translational studies using circulating tumor DNA (ctDNA) and comprehensive genomic profiling help elucidate resistance mechanisms, providing opportunities to adjust therapy in real time and guide future drug development.

Future Directions and Challenges

Emerging Trends
Overall, the future directions in drug development for HR+ HER2– breast cancer are marked by several overlapping trends:
• A growing emphasis on the development of novel endocrine agents that are more potent, orally bioavailable, and capable of triggering receptor degradation regardless of mutation status. These next-generation SERDs and dual-modulating agents represent a significant trend in clinical research.
• There is an increasing focus on combination therapies that simultaneously target multiple pathways driving endocrine resistance. Combining endocrine therapy with targeted agents such as CDK4/6 inhibitors, PI3K inhibitors, and FGFR inhibitors is rapidly becoming a standard clinical research approach in this setting.
• The integration of immunotherapy into the HR+ treatment algorithm is emerging as another frontier. Although the immune microenvironment in HR+ breast cancer has been traditionally regarded as less inflamed, research that is “re-warming” the tumor immune milieu with combined targeted and immunomodulatory agents is an exciting trend that may have future clinical ramifications.
• Personalized medicine approaches, which incorporate genomic and transcriptomic profiling, are increasingly used to guide clinical trial design. These methods identify actionable mutations (such as ESR1, PIK3CA, and FGFR alterations) that can be targeted by novel drugs, thereby tailoring treatment to individual tumor biology.

Challenges in Drug Development
Despite the promising advances, several critical challenges remain in the development of new drugs for HR+ HER2– breast cancer:
• Endocrine resistance mechanisms are multifactorial, involving genomic alterations, epigenetic modifications, and complex feedback loops in intracellular signaling. Successfully targeting this multifaceted resistance requires drugs that have multi-target properties or the rational combination of multiple agents.
• The safety profiles of combination regimens can be challenging. Many targeted agents, while efficacious in preclinical models, have shown overlapping toxicities such as hyperglycemia (for PI3K inhibitors) or myelosuppression (with CDK4/6 inhibitors). Designing regimens that optimize efficacy while minimizing toxicity remains a delicate and ongoing challenge.
• Heterogeneity of HR+ HER2– tumors at the molecular level is significant. This heterogeneity complicates patient selection and response prediction. Despite advances in biomarker identification, there is still a need for validated predictive markers that can reliably assess which patients will benefit from a given therapy.
• Regulatory challenges and the need for long-term clinical endpoint data are further hurdles. As many agents are in early-phase studies, large phase III trials are necessary to validate their clinical benefit, especially when comparing novel combinations to established endocrine-based therapies.
• Finally, integrating immunotherapy in a “traditionally non-immunogenic” tumor type poses additional problems. It is not yet clear if the synergy observed in combination trials will translate into meaningful long-term benefits, and if the modulation of the immune microenvironment can overcome established hormone resistance.

Conclusion
In summary, the development of new drugs for hormone receptor–positive, HER2–negative breast cancer is a multi-pronged effort targeting several resistance mechanisms that limit the efficacy of current endocrine therapies. Novel hormone therapies, such as next-generation oral SERDs and dual-action endocrine agents, are being produced to ensure more complete blockade of estrogen signaling—even in the presence of ESR1 mutations. In the realm of targeted therapies, next-generation PI3K/AKT/mTOR inhibitors, new CDK4/6 inhibitors with optimized toxicity profiles, and FGFR inhibitors are all under active evaluation in early-phase and combination clinical trials. Furthermore, although immunotherapy has not historically been a mainstay in HR+ breast cancer treatment, emerging immune-based approaches—whether checkpoint inhibitors, therapeutic vaccines, or adoptive cell therapies—are being explored to enhance response rates and overcome resistance.

Ongoing clinical trials are leveraging biomarker-driven patient selection to ensure that therapies are tailored to the unique molecular profiles of tumors, and recent research findings consistently highlight the benefit of multi-agent approaches that block multiple resistance pathways. The future of drug development in this setting is promising, yet challenges remain regarding optimal patient selection, toxicity management, and overcoming tumor heterogeneity. Continued translational research and well-designed clinical trials are essential to validate these emerging agents and integrate them into clinical practice effectively.

In conclusion, the drugs in development for HR+ HER2– breast cancer represent a dynamic and diverse portfolio of novel endocrine, targeted, and immunotherapeutic strategies that aim to improve outcomes by addressing the underlying molecular vulnerabilities and mechanisms of resistance. While significant progress has been made, further research is needed—as evidenced by numerous ongoing trials—to ensure that the challenges of resistance, toxicity, and patient heterogeneity can be overcome. Ultimately, these innovative approaches promise a future where treatment is personalized, multi-pronged, and capable of offering long-term disease control to patients with this prevalent subtype of breast cancer.

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.