What is the mechanism of action of Inavolisib?

7 March 2025

Introduction to Inavolisib

Inavolisib is an innovative, investigational oral targeted therapy that has emerged as a novel approach in the treatment of certain forms of cancer, most notably hormone receptor (HR)-positive, HER2-negative advanced breast cancer harboring PIK3CA mutations. Structurally classified as a small molecule drug, it represents a first‐in‐class agent that distinguishes itself by not only binding to its primary target but also by promoting the degradation of the mutant protein responsible for aberrant signaling. In preclinical studies and early-phase clinical trials, inavolisib demonstrated potent in vitro activity and robust anti-tumor effects, making it a candidate for relieving the burden of resistance that often develops when conventional endocrine therapies fail. The drug has received considerable attention due to its dual mode of action—both enzymatically inhibiting a key signaling pathway and mediating the downregulation of mutant oncoproteins—thus offering a more tailored therapeutic strategy compared to more general PI3K inhibition methods. Furthermore, its oral bioavailability and manageable pharmacokinetic profile (achieving steady state by Day 5 and exhibiting approximately two-fold accumulation) add to its attractiveness as a component of combination regimens for bulky and difficult-to-treat tumors.

Therapeutic Uses

Therapeutically, inavolisib is primarily being developed for patients with HR-positive, HER2-negative metastatic breast cancer that exhibit mutations in the PIK3CA gene. These mutations frequently result in a constitutively active PI3Kα protein that drives tumor progression, conferring resistance to endocrine-based treatment and leading to poorer clinical outcomes. In clinical trials, inavolisib is commonly administered in combination with other established agents such as palbociclib—a cyclin-dependent kinase inhibitor—and fulvestrant, an estrogen receptor antagonist, to maximize its therapeutic benefit. The rationale behind the combination is rooted in the observation that while palbociclib and fulvestrant target cell cycle progression and estrogen receptor signaling respectively, inavolisib specifically intercepts the overactive PI3K/AKT pathway resulting from the PIK3CA mutation. With regulatory advances marked by its approval in the United States on October 10, 2024, the drug has rapidly ascended the clinical development ladder, with multiple Phase III clinical studies (e.g., INAVO120, INAVO121, and INAVO122) ongoing or completed to further elucidate its efficacy and safety profile.

Mechanism of Action

The mechanism of action of inavolisib can be dissected into multiple facets revolving around its molecular target specificity and its influence on major biochemical pathways within the cell.

Molecular Targets

At its core, inavolisib is specifically designed to inhibit the enzymatic activity of phosphatidylinositol 3-kinase alpha (PI3Kα), which is encoded by the PIK3CA gene. PI3Kα is a vital enzyme involved in the transduction of growth, proliferation, and survival signals in various cells. Importantly, in many cancers, particularly breast cancers harboring PIK3CA mutations, the PI3Kα protein becomes constitutively active, leading to uncontrolled cell growth and resistance to conventional endocrine therapies. The agent binds with high affinity selectively to the catalytic subunit p110α of PI3Kα and not merely blocks it at the active site; it uniquely induces the degradation of mutated p110α. This dual action—both inhibition and degradation—is crucial for diminishing the excessive signaling that promotes cancer cell proliferation. Because the mutant form of p110α is predominantly responsible for driving tumor progression, inavolisib’s preferential interaction with the mutant protein represents a significant advancement compared to other PI3K inhibitors that may target the wild-type enzyme as well, thereby avoiding collateral inhibition of normal cellular processes. This contributes to its favorable tolerability profile, aligning with the goal of precision medicine. Beyond its primary target, there is emerging data suggesting that inavolisib might modulate ancillary signaling nodes indirectly by altering the dynamics of downstream effectors in the PI3K pathway, although its principal and most impactful interaction remains with PI3Kα.

Biochemical Pathways

The PI3K/AKT signaling pathway is central to many cellular functions such as metabolism, growth, proliferation, and survival. Inavolisib disrupts this pathway in several important ways: Once PI3Kα is activated by receptor tyrosine kinases (RTKs) or other upstream signals, it normally phosphorylates phosphatidylinositols to generate PIP3, which in turn recruits AKT to the cell membrane. Activation of AKT then leads to a cascade that promotes cellular survival and proliferation. Inavolisib, by inhibiting PI3Kα, prevents the formation of PIP3 and the consequent activation of AKT. This blockade leads to a reduction in the phosphorylation of downstream targets—including mTOR and other kinases—which are essential for driving the cell cycle and tumor cell survival. Importantly, inavolisib’s mechanism is not linear inhibition alone; its ability to trigger the degradation of the mutant PI3Kα protein suggests a prolonged and robust termination of the aberrant signaling process. This destruction of the oncoprotein removes not just the catalytic activity but also reduces the scaffold functions that the mutant protein may exert within oncogenic signaling complexes. The disruption of the PI3K/AKT pathway by inavolisib results in several downstream cellular events: there is a decrease in anti-apoptotic signals and a concomitant increase in pro-apoptotic factors, ultimately tipping the balance towards cell death in tumor cells that rely heavily on this survival pathway. The inhibition also affects translation and metabolism by interfering with mTOR complex 1 signaling, further hindering cell growth and protein synthesis. Moreover, the inhibition of PI3Kα has been shown to reduce cellular proliferation through the modulation of cell cycle regulators. Arrest in certain phases of the cell cycle (specifically the G1 to S phase transition) has been noted in preclinical models, providing additional mechanistic insight into how tumor growth is curtailed upon exposure to inavolisib.

Effects on Cellular Processes

The biochemical interventions mediated by inavolisib have profound effects both on cancer cells and, to a lesser degree, on normal cells. These outcomes are a reflection of the drug’s specificity and its ability to disrupt critical cellular processes that drive malignancy.

Impact on Cancer Cells

Cancer cells with PIK3CA mutations exhibit an over-reliance on the PI3K/AKT pathway for growth, survival, and resistance to stress signals. Inavolisib exploits this dependency in several distinct manners: By inhibiting PI3Kα and promoting degradation of its mutant form, inavolisib effectively reduces the constitutive activation of AKT. This reduction in AKT activation leads to a downstream inhibition of mTOR signaling, which is crucial for protein synthesis and cellular metabolism. The cumulative effect is a marked reduction in the proliferative capacity of cancer cells. The suppression of survival signaling pathways results in an induction of programmed cell death (apoptosis). Cancer cells undergoing treatment with inavolisib display increased apoptotic markers as a consequence of diminished pro-survival signals coupled with the activation of caspase cascades—a hallmark of the intrinsic apoptotic pathway. This apoptotic induction is particularly pronounced in tumor cells that have adapted to rely on the mutated PI3Kα-driven pathway for their longevity. Inavolisib also disrupts cell cycle progression. In many in vitro experiments, cells treated with the drug show evidence of cell cycle arrest that impedes the transition from the G1 to S phase. This arrest not only limits the replication potential of the tumor cells but also sensitizes the cells to the effects of combination therapy with agents such as palbociclib (which directly targets cell cycle progression) and fulvestrant (which impairs estrogen receptor-mediated survival). Furthermore, the highly selective inhibition offered by inavolisib allows it to target cancer cells while sparing many of the normal proliferative pathways in non-tumor cells, a factor that ultimately contributes to reduced systemic toxicity. Preclinical data support that the drug’s efficacy is closely tied to the presence of PIK3CA mutations, meaning that its tumoricidal effects are most robust in cells that harbor this biomarker.

Effects on Normal Cells

In normal cells where the PI3K pathway functions under tightly regulated conditions, the impact of inavolisib is less severe compared to its effects on cancer cells with mutant PIK3CA: Given that non-malignant cells typically do not have a constitutively active PI3Kα pathway, the inhibition by inavolisib does not result in the same degree of pathway suppression. Therefore, the levels of AKT phosphorylation in these cells are only modestly affected, allowing normal cell growth, survival, and metabolic functions to continue with minimal disruption. Clinical trial data and pharmacokinetic studies have confirmed that, at recommended doses, inavolisib exhibits a manageable safety profile. The most common drug-related adverse events observed in trials (e.g., hyperglycemia, neutropenia, stomatitis) are reflective of the on-target effects on systemic metabolism and immune cell proliferation rather than overt toxicity resulting from the inhibition of PI3K in normal tissues. The specificity for mutant PI3Kα is a critical factor that reduces the incidence of off-target side effects. The fact that inavolisib does not significantly impair other isoforms of PI3K allows normal cells to maintain adequate signaling through less critical or redundant pathways. This targeted action minimizes potential collateral damage and ensures that normal physiological processes are largely preserved, contributing to a better tolerability profile in patients undergoing treatment. On a molecular level, while in normal cells some inhibition of the baseline PI3K activity occurs, the extent of this inhibition is not sufficient to derail the cascade of downstream signals required for routine cellular functions. Instead, the selective pressure on cancer cells that are addicted to the hyperactive pathway is significantly higher, leading to differential outcomes between malignant and non-malignant tissues.

Clinical Implications and Research

The detailed understanding of inavolisib’s mechanism of action has extensive clinical implications and inspires future research to further enhance and optimize its use as part of combination treatments or as a monotherapy in certain cancer subtypes.

Clinical Trial Results

Clinical trials involving inavolisib, particularly the Phase III INAVO120 study, have provided compelling evidence for its efficacy when combined with established agents like palbociclib and fulvestrant. In these studies: Patients with PIK3CA-mutated, HR-positive, HER2-negative metastatic breast cancer achieved a significant clinical benefit. The inavolisib combination reduced the risk of progression by approximately 57% compared to standard treatment, with a median progression-free survival (PFS) of 15.0 months versus 7.3 months in the comparator arm. Secondary endpoints such as objective response rate, duration of response, and clinical benefit rate further underscore the drug’s impact when added to combination regimens. These outcomes suggest that inavolisib’s action in curtailing the PI3K/AKT pathway not only delays disease progression but also contributes to tumor shrinkage and sustained disease control. The clinical trial data also indicate that the adverse events associated with inavolisib, such as hyperglycemia and neutropenia, are consistent with its on-target effects, yet they remain clinically manageable. The tolerability outcomes reinforce that the selective inhibition of the PI3Kα isoform is largely confined to malignant cells while sparing normal tissues. These robust clinical results, in conjunction with its dual mechanism of both enzymatic inhibition and protein degradation, position inavolisib as a potentially transformative treatment option for patients with advanced PIK3CA-mutated breast cancer. Moreover, its unique mode of action provides a scientific rationale for its use not only in breast cancer but possibly in other malignancies driven by PI3K dysregulation.

Future Research Directions

Despite the significant progress made in understanding inavolisib’s mechanism of action and its clinical benefits, several avenues remain open for further research and optimization: Further elucidation of the molecular basis of inavolisib-induced degradation of mutant p110α is needed. Advanced studies employing proteomic analyses and time-course experiments in diverse tumor models could provide deeper insights into the kinetics and regulatory pathways involved in this process. Investigating biomarkers that could predict or correlate with response to inavolisib is another important research direction. Although PIK3CA mutation status is a primary inclusion criterion, additional markers (such as downstream signaling components, expression of anti-apoptotic proteins, or alterations in cell cycle regulators) may refine patient selection and maximize therapeutic benefits. Expanding the scope of inavolisib beyond breast cancer to other tumor types with aberrant PI3K/AKT signaling is of considerable interest. Preclinical studies have begun to validate its efficacy in other solid tumors, and future clinical trials might assess combination therapies in different oncological settings. Moreover, long-term studies are warranted to evaluate the durability of response with inavolisib-based therapy, potential acquired resistance mechanisms, and the long-term safety of inhibiting a pathway that is also involved in normal cellular functions. Such studies will inform dose adjustment, treatment duration, and strategies to mitigate resistance. Research into combination strategies is also critical. Combinations of inavolisib with other targeted agents (such as CDK inhibitors like palbociclib, endocrine therapies like fulvestrant, or even novel immunotherapeutic approaches) necessitate a deeper understanding of how simultaneous inhibition of multiple signaling axes might produce synergistic anti-tumor effects while preserving a tolerable side effect profile. Additionally, there is potential for inavolisib to be integrated into treatment protocols that address the heterogeneity of cancer cells within solid tumors. Understanding the interplay between tumor microenvironment factors, immune modulators, and PI3K signaling inhibition may lead to innovative approaches to overcome drug resistance and improve patient outcomes.

Conclusion

In summary, the mechanism of action of inavolisib is a multifaceted process that involves highly selective inhibition of phosphatidylinositol 3-kinase alpha (PI3Kα), a key driver of tumorigenesis in PIK3CA-mutated cancers. Its primary function is to bind to and inhibit the catalytic activity of the mutant p110α subunit of PI3K, thereby impeding the formation of phosphatidylinositol-(3,4,5)-trisphosphate (PIP3) and preventing subsequent activation of AKT and downstream effectors involved in cell survival, proliferation, and protein synthesis. Beyond simple enzymatic inhibition, inavolisib uniquely induces the degradation of the mutant protein, ensuring a sustained suppression of the oncogenic PI3K/AKT/mTOR pathway.

On a cellular level, these molecular events translate into a reduction in tumor cell proliferation, the induction of apoptosis through intrinsic cell death pathways, and the disruption of cell cycle progression. These effects are particularly pronounced in cancer cells that depend heavily on the aberrant signaling driven by PIK3CA mutations, while normal cells are relatively spared due to their reliance on the untampered, well-regulated PI3K isoforms. Clinically, the promising outcomes reported in major trials—such as the marked extension of progression-free survival in the INAVO120 study—underscore the therapeutic value of this targeted approach and provide a rationale for combining inavolisib with other anticancer regimens.

Looking forward, ongoing research is aimed at deepening our understanding of the drug’s mechanism, identifying biomarkers of response, and expanding its application to other cancers with activated PI3K signaling. The future of inavolisib is tied to not only optimizing combination strategies but also to addressing resistance mechanisms and ensuring long-term safety. Together, these facets establish inavolisib as a promising and transformative agent in the era of precision oncology, offering new hope for patients with difficult-to-treat malignancies while maintaining an emphasis on preserving quality of life through a manageable safety profile.

In conclusion, inavolisib’s dual ability to inhibit and degrade mutant PI3Kα represents a significant advancement in targeted cancer therapy. Its multi-angle approach—spanning molecular targeting, biochemical pathway modulation, and favorable clinical outcomes—provides a strong foundation for its continued development and integration into individualized treatment strategies for patients with advanced breast cancer and potentially other tumor types where the PI3K pathway is aberrantly activated.

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.