Introduction to USP7
USP7, also known as
HAUSP (herpesvirus-associated ubiquitin-specific protease), is one of the most widely studied
deubiquitinating enzymes (DUBs) in the human ubiquitin–proteasome system. A major focus of
cancer research and increasingly of other disease areas, USP7 modulates the protein half-life of many substrates that control cell survival, genome stability, and immune functions. Its complex and multi-domain structure—including a
TRAF-like domain, the central catalytic domain, and ubiquitin-like (UBL) regions linked via a flexible connector—ensures that USP7’s deubiquitinating activity is tightly regulated and only activated upon binding to ubiquitin.
Biological Role and Mechanism
USP7’s primary biological role is the removal of ubiquitin chains from substrate proteins and prevention of their proteasomal degradation. This process has far-reaching consequences for pathways such as the
p53–
MDM2 axis, DNA damage repair, cell cycle progression, epigenetic regulation, and immune signaling. Notably, USP7 deubiquitinates both p53, a crucial tumor suppressor, and MDM2, the E3 ligase that targets p53 for degradation. Under physiological conditions, USP7’s balanced action contributes to protein homeostasis. However, USP7 is sensitive to allosteric regulation; its catalytic triad remains in an “inactive” state until conformational changes induced by ubiquitin engagement precisely position its active residues. This unique mechanism explains why targeting USP7 with inhibitors is challenging yet potentially transformative, given the enzyme’s switch-like activation mode during cellular stress.
Importance in Disease Pathology
The importance of USP7 in disease becomes evident when its regulation fails. Abnormal or overexpression of USP7 has been linked to multiple human cancers as it stabilizes oncogenic proteins (e.g. MDM2) and contributes to the inactivation of p53 through complex feedback loops. Also, USP7 is implicated in genomic instability, improper DNA damage response, and epigenetic alterations that support
tumorigenesis. Beyond cancer, emerging evidence suggests a role for USP7 in immunomodulation and even neurodegenerative disorders by altering the stability of proteins that regulate immune checkpoint molecules or neuronal survival signals. In addition, recent studies have tied USP7 to the regulation of T regulatory cells (Tregs) and chromatin modifiers such as EZH2. Therefore, the dysregulation of USP7 constitutes a common denominator in pathways underpinning several pathologies, making it an ideal target for drug discovery.
USP7 Inhibitors
USP7 inhibitors have rapidly evolved from early micromolar compounds to more potent, highly selective next-generation molecules that show promise in preclinical models. The discovery and optimization of these inhibitors have been essential not only for validating USP7 as a therapeutic target but also for enabling interventions in complex signaling networks.
Mechanism of Action
USP7 inhibitors typically function by interfering with the enzyme’s catalytic activity and substrate binding. Some of these inhibitors covalently modify the catalytic cysteine (C223) of USP7, leading to irreversible inactivation and sustained downstream biological effects such as decreased MDM2 stabilization and restoration of p53 levels. In contrast, other inhibitors bind allosterically, stabilizing an inactive conformation of USP7 and preventing ubiquitin binding. This dual strategy—covalent versus non-covalent (allosteric) inhibition—provides an array of approaches for different therapeutic settings. For example, covalent inhibitors like P5091 and its optimized derivatives (e.g. P22077) have demonstrated the ability to trigger apoptosis in tumor cells by disrupting the USP7–MDM2–p53 network. In addition, allosteric inhibitors have the potential to offer better selectivity and reversible binding properties, thereby reducing potential off-target effects.
Development and Types
Over the last decade, numerous small molecules have been developed that target USP7. Early inhibitors such as HBX-41108 and P5091 provided the initial proof of concept that USP7 inhibition could stabilize p53 and impair tumor cell viability. Subsequent refinement of these molecules led to improved analogs such as FT671, FT827, and compound 4, characterized by nanomolar potency, enhanced selectivity, and favorable pharmacokinetic properties. Recent structure-guided approaches have further improved the specificity of USP7 inhibitors, often by exploiting previously undisclosed binding pockets distant from the catalytic cleft. The diversity in chemical scaffolds—ranging from irreversible covalent binders to reversible allosteric inhibitors—has created a robust drug discovery pipeline, enabling researchers to choose the most appropriate molecule based on the therapeutic context and desired pharmacological profile.
Therapeutic Applications
The therapeutic applications of USP7 inhibitors are diverse, with cancer treatment being the most comprehensively studied area. Nonetheless, there is growing interest in the potential application of these inhibitors in neurodegenerative diseases and other conditions. Below are the detailed potential applications:
Cancer Treatment
Cancer is the most widely studied field for USP7 inhibitor application. Due to its central role in the p53–MDM2 axis, USP7 inhibitors can restore p53 function through MDM2 destabilization. This is particularly important in tumors where p53 is wild type but functionally suppressed by enhanced MDM2 activity. In multiple myeloma models, for example, the USP7 inhibitor P5091 has been shown to effectively reduce tumor growth by triggering p53-mediated apoptosis.
Besides hematologic malignancies, studies have demonstrated anticancer activity in solid tumors such as colon, prostate, and lung cancers. The inhibition of USP7 not only affects p53 but also destabilizes other substrates that drive oncogenesis, including FOXO and EZH2. Moreover, combinatorial approaches have shown that pairing USP7 inhibitors with agents targeting PLK1 (a key mitotic kinase) or DNA-damaging chemotherapeutics increases cell death in taxane-resistant cancer cells. This synergy suggests a promising combination therapy strategy for addressing chemo-resistance issues, which have been a major challenge in current oncology practices. Furthermore, USP7’s involvement in immune modulation, particularly in T regulatory cells (Tregs), offers the possibility of integrating USP7 inhibitors with cancer immunotherapies, potentially enhancing anti-tumor immunity. These multi-faceted anti-cancer effects make USP7 inhibitors promising candidates with potentially broad applications across several cancer types.
Neurodegenerative Diseases
Beyond oncology, emerging research suggests USP7 inhibition might have therapeutic implications in neurodegenerative disorders. Although the primary focus to date has been on cancer, studies hint at USP7’s role in neuronal homeostasis. USP7 regulates substrates that are important not only for cell cycle control and apoptosis but also for maintaining protein quality control in neurons. Neurodegenerative diseases, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD), are characterized by the accumulation of misfolded proteins and dysfunctional cellular clearance mechanisms.
The restoration of proteostasis by modulating the ubiquitin–proteasome system has been an attractive strategy in neurodegeneration research. In particular, USP7 inhibitors may enhance the clearance of toxic proteins by altering the balance between protein ubiquitination and degradation. Furthermore, a recent study on the USP7 inhibitor P5091 and next-generation molecules suggests that by deubiquitinating proteins involved in neuronal survival, USP7 inhibition might provide neuroprotective effects. These findings indicate that although still in early preclinical phases, targeting USP7 could be valuable for diseases characterized by protein aggregation and cell death in the central nervous system. In addition, because USP7 has been linked to the regulation of immune responses within the brain (for instance, by affecting Tregs and microglia function), its inhibition could indirectly modulate neuroinflammation—a common pathology in neurodegenerative disorders.
Other Potential Applications
Aside from cancer and neurodegeneration, the pharmacological modulation of USP7 could have applications in other areas where the ubiquitin–proteasome system plays a critical role. For instance, USP7 inhibitors are being investigated for their potential in regulating immune responses. Given the enzyme’s role in stabilizing proteins that modulate the immune checkpoint (such as PD-L1 via indirect pathways), USP7 inhibition carries the prospect of enhancing anti-tumor immune responses and may find applications in autoimmune disorders.
In addition, USP7’s broad substrate repertoire hints at roles in DNA damage repair and cell stress responses. As such, inhibitors might be used in combination with conventional therapies to enhance the sensitivity of tumors to radiotherapy or chemotherapy. Some preclinical models, for example, indicate that the combination of USP7 inhibitors with DNA-damaging agents leads to increased tumor cell apoptosis, making them potential adjuvant treatments in oncology. Moreover, further exploration might reveal therapeutic applications in conditions characterized by senescence or age-related pathologies, as USP7 inhibition has been linked to the selective elimination of senescent cells via the restoration of p53 activity. Although these applications are at an early stage of exploration, they open new avenues for the treatment of diverse diseases beyond the classical oncology realm.
Research and Clinical Trials
Extensive preclinical research supports the promise of USP7 inhibitors in multiple therapeutic areas, and several clinical studies are on the horizon to validate these findings.
Current Research Findings
Much of the current state-of-the-art research on USP7 inhibitors comes from in vitro and in vivo studies conducted in diverse cancer models. Research has consistently shown that USP7 inhibition leads to destabilization of MDM2, thereby increasing p53 activity and promoting apoptosis in various tumor cell lines. For example, treatment with USP7 inhibitors like P5091 and its analogs in multiple myeloma xenograft models has resulted in significant tumor suppression and improved survival outcomes.
Furthermore, structure-based drug design has yielded potent, nanomolar-range inhibitors that bind either covalently or allosterically to USP7, providing detailed insights into the binding pockets and the dynamic conformational changes triggered by inhibitor binding. These studies are supported by crystallographic data and molecular dynamics simulations that offer a mechanistic rationale for the observed cellular phenotypes upon USP7 inhibition.
In addition to traditional anti-cancer effects, emerging research has begun to document the neuroprotective potential of USP7 inhibition. Preclinical data indicate that USP7 inhibitors can modulate pathways involved in proteostasis and neuroinflammation, thereby reducing neuronal damage and improving behavioral outcomes in animal models of neurodegeneration. Complementing these studies, evidence also suggests that USP7 inhibitors may modulate the immune microenvironment by impairing Treg functions, thereby contributing not just to direct tumor cell killing but also enhancing immunotherapy responses.
Collectively, these studies illustrate that USP7 inhibitors act on multiple signaling axes, ranging from direct tumor suppression (via modulation of p53/MDM2) to immune system modulation and even protein quality control in neurodegeneration. This body of work supports the rationale for advancing USP7 inhibitors into clinical evaluation.
Ongoing Clinical Trials
Although most USP7 inhibitors remain in the preclinical phase, the momentum of research is driving early-stage clinical investigations. Several clinical trial programs are evaluating the safety, tolerability, and preliminary efficacy of these compounds in select cancers where the USP7 pathway is dysregulated. For instance, while compounds like P5091 and its derivatives have entered advanced preclinical testing, second-generation inhibitors (such as FT671) are being optimized for oral bioavailability and low toxicity in preparation for first-in-human trials.
Moreover, investigators are examining the potential of combining USP7 inhibitors with existing chemotherapeutics or targeted agents to overcome drug resistance, based on compelling preclinical synergy data. In the realm of immuno-oncology, some trial designs intend to test combinations of USP7 inhibitors with immune checkpoint inhibitors, hoping to further harness the immune-modulating effects of USP7 inhibition.
These early clinical investigations are guided by robust preclinical evidence and structural insights that promise improved specificity and safety. As these studies progress, outcomes such as tolerance profiles, pharmacodynamics markers (e.g. MDM2 degradation and p53 upregulation), and even effects on regulatory T cell populations will be critical endpoints to validate the therapeutic potential of USP7 inhibitors.
Challenges and Future Directions
Even though the therapeutic promise of USP7 inhibition is extensive, several significant challenges must be addressed to fully realize their potential in a clinical setting.
Drug Development Challenges
First, the intrinsic regulatory complexity of USP7 is itself a challenge. Because its catalytic activity is only triggered upon conformational rearrangement following ubiquitin binding, designing inhibitors that can precisely target the active or relevant allosteric sites without affecting other DUBs requires high structural resolution and exquisite medicinal chemistry. Early inhibitor candidates like HBX-41108 exhibited issues with selectivity and off-target effects, underscoring the need for molecules that not only inhibit USP7 effectively but also spare other deubiquitinating enzymes.
Second, pharmacokinetic challenges such as achieving sufficient bioavailability, proper tissue distribution, and metabolic stability remain critical hurdles. Ensuring that the inhibitors reach effective concentrations at the tumor site (or in the brain in case of neurodegenerative disorders) while minimizing systemic toxicity requires rigorous optimization of chemical scaffolds. Resistance mechanisms may also emerge, akin to other targeted therapies. Hence, ongoing research must anticipate potential resistance pathways—such as mutations in the USP7 binding site or compensatory upregulation of parallel deubiquitinating enzymes—and consider combination therapeutic strategies to counteract these effects.
Finally, another drug development obstacle is the intrinsic complexity of the downstream pathways regulated by USP7. Because USP7 has multiple substrates that influence diverse cell functions, inhibitors might produce unintended effects in normal tissues. For instance, while reactivating p53 is beneficial in tumor cells, the same mechanism could potentially disturb tissue homeostasis in non-cancerous cells. Such pleiotropic effects necessitate careful dose optimization and patient selection strategies in clinical trials to achieve a favorable therapeutic index.
Future Research Directions
Future research on USP7 inhibitors is poised to focus on three main avenues:
1. Refinement of Inhibitor Design: Further exploration of allosteric sites and covalent–non-covalent binding modalities using advanced computational methods, nuclear magnetic resonance (NMR) spectroscopy, and high-resolution crystallography will continue to provide crucial insights. These structural studies will guide the development of next-generation inhibitors with improved potency, selectivity, and pharmacokinetic profiles.
2. Expanded Therapeutic Indications: While cancer remains the paramount indication for USP7 inhibition, studies probing its role in neurodegeneration and immune modulation are rapidly expanding. Future investigations should include comprehensive animal models to confirm the neuroprotective effects and explore whether USP7 inhibition can modulate the immune microenvironment in transplantable or spontaneous tumor models. Additionally, research into the role of USP7 in senescence and its connection with aging-related diseases could widen its therapeutic applicability beyond oncology.
3. Clinical Translation and Biomarker Development: As clinical trials using USP7 inhibitors advance, extensive biomarker studies will be imperative to select patients who will benefit most from such therapies. Biomarkers could include the expression levels of USP7, MDM2, p53, and other substrates as well as immune markers such as T regulatory cell counts. Moreover, the success of combination therapies with chemotherapy, kinase inhibitors, or immunotherapies will require careful evaluation, from preclinical models through to clinical phases, to ensure the optimal harnessing of USP7 inhibition’s multi-pronged mechanism of action.
Additionally, investigating the interplay between USP7 inhibitors and other drugs targeting the ubiquitin–proteasome system (such as proteasome inhibitors and other specific DUB inhibitors) may facilitate the design of combination regimens that address tumor drug-resistance and clinical heterogeneity. Parallel efforts are needed to evaluate potential toxicities in normal tissues and to establish dosing schedules that maximize efficacy while minimizing adverse effects.
Conclusion
In summary, USP7 is a critical regulator of cellular homeostasis, impacting pivotal proteins such as p53, MDM2, EZH2, and factors involved in DNA repair and immune regulation. The targeted inhibition of USP7 has been shown to restore tumor suppressor functions, induce apoptosis in cancer cells, and even modulate immune responses by interfering with T regulatory cell function. With the development of various inhibitors—from early covalent binders to sophisticated allosteric molecules—researchers have generated potent compounds such as P5091, P22077, FT671, and others that effectively modulate USP7 activity in preclinical models.
Therapeutically, the applications for USP7 inhibitors center primarily on cancer treatment, where restored p53 function and MDM2 destabilization promote tumor cell apoptosis and even overcome drug-resistance mechanisms when used in combination with standard chemotherapeutics. Additionally, emerging data support the potential use of USP7 inhibitors in neurodegenerative disorders, where the restoration of protein homeostasis and reduction of neuroinflammation may yield neuroprotective benefits. Other potential cutting-edge applications include immunomodulation, where USP7 inhibition could enhance responses to immunotherapies, and the treatment of age-related diseases by selectively targeting senescent cells.
Current research findings underscore both the mechanistic insights and the promising anticancer and neuroprotective outcomes of USP7 inhibition, with early-stage clinical trials under development to validate these findings in patients. However, challenges remain in the form of selectivity, potential off-target effects, pharmacokinetic hurdles, and the inherent complexity of USP7’s cellular roles. Looking forward, future directions will likely emphasize further refinement of inhibitor design, expansion into new therapeutic indications, and robust clinical trial programs augmented by biomarker-driven patient selection.
In conclusion, USP7 inhibitors represent a transformative approach in targeted therapy with significant potential for treating diverse diseases—from aggressive cancers resistant to current therapies to neurodegenerative disorders marked by proteotoxicity. Continued research, advanced chemical optimization, and carefully designed clinical studies are crucial to fully harness the therapeutic promise of USP7 inhibition, paving the future for personalized and combinatorial treatments that may ultimately change the standard of care in oncology and beyond.