ZB-004, also identified by the research designation XmAb10717, is an investigational biopharmaceutical agent currently under clinical development.[1] It is a bioengineered fusion protein, specifically classified as a cytotoxic T-lymphocyte-associated antigen 4‑immunoglobulin (CTLA-4-Ig).[3] Functionally, ZB-004 acts as an immunomodulator by selectively inhibiting the costimulatory signals essential for T-lymphocyte activation, a pivotal process in the adaptive immune response.[2] The primary therapeutic focus for ZB-004 is the treatment of autoimmune diseases, conditions wherein aberrant T-cell activity contributes significantly to pathogenesis.[1]
ZB-004 represents a second-generation therapeutic within the CTLA-4-Ig class. This classification is underscored by its design, which incorporates specific bioengineering modifications aimed at optimizing its pharmacological profile relative to earlier CTLA-4-Ig molecules, such as abatacept. The explicit design goals for ZB-004 include "increased binding affinity" to its target ligands (CD80 and CD86) and an "extended half-life".[3] These enhancements are intended to build upon the clinically validated mechanism of action of CTLA-4-Ig [7] by potentially offering superior pharmacological properties. Such improvements could manifest as enhanced efficacy, possibly due to more potent target engagement, and improved patient convenience through a less frequent dosing schedule.
The conception and initial development of ZB-004 (XmAb10717) were undertaken by Xencor, Inc. This development leveraged Xencor's proprietary XmAb® antibody engineering platform, with a key component being the Xtend™ Fc technology, specifically designed to prolong the circulating half-life of therapeutic proteins.[2]
In November 2020, Xencor, Inc. executed a licensing agreement that granted exclusive worldwide rights for the continued development and subsequent commercialization of XmAb10717 (ZB-004), along with two other preclinical assets (XmAb6755 and XPro9523), to a newly formed, privately held biotechnology company.[9] The entities currently spearheading the clinical advancement of ZB-004 are Zenas BioPharma LLC and its affiliated company, Shanghai Zenas Biotechnology Co., Ltd..[2] This transition in stewardship occurred while ZB-004 was in the preclinical phase of development, with Zenas BioPharma subsequently guiding the molecule into Phase 1 clinical evaluation.[2]
The transfer of ZB-004's development from Xencor to Zenas BioPharma illustrates a common strategic approach within the biopharmaceutical sector. Xencor, as a company with a strong focus on technology platforms and early-stage drug discovery, excels in innovating and de-risking novel therapeutic candidates through advanced protein engineering. Their business model frequently involves out-licensing or partnering programs after initial validation.[9] Conversely, Zenas BioPharma was established with a specific mission to undertake later-stage clinical development and global commercialization of immune-based therapies. The acquisition of assets like ZB-004 aligns with Zenas' strategy to construct a robust pipeline by securing promising, engineered molecules that possess a strong scientific rationale and the potential for significant clinical differentiation.[2] This division of labor allows each organization to capitalize on its core competencies, potentially streamlining and accelerating the overall development trajectory of ZB-004.
Table 1: ZB-004 (XmAb10717) - Key Characteristics
Feature | Description | References |
---|---|---|
Synonyms | ZB004, ZB 004, XmAb10717 | 2 |
Drug Class | CTLA-4-Ig Fusion Protein, Immunomodulator | 2 |
Originator | Xencor, Inc. | 2 |
Current Developer | Zenas BioPharma LLC / Shanghai Zenas Biotechnology Co., Ltd. | 2 |
Molecular Target | CD80 (B7-1) and CD86 (B7-2) on antigen-presenting cells | 3 |
Key Engineering Features | CTLA-4 Extracellular Domain (ECD) substitutions for increased CD80/CD86 binding affinity | 3 |
Xtend™ Fc Domain (hybrid IgG1/2 Fc region with M428L and N434S substitutions) for extended plasma half-life | 3 |
ZB-004 is a sophisticated, bioengineered recombinant fusion protein. Its fundamental architecture involves the genetic fusion of the extracellular domain (ECD) of human Cytotoxic T-Lymphocyte-Associated Antigen 4 (CTLA-4) with a modified human immunoglobulin (Ig) Fc domain.[3] CTLA-4 is a critical natural negative regulator of T-cell immune responses. By presenting the ligand-binding portion of CTLA-4 in a soluble, stable, and pharmacokinetically optimized format, ZB-004 is designed to mimic and amplify this natural inhibitory function.[3] The therapeutic principle of CTLA-4-Ig fusion proteins is well-established in the treatment of autoimmune diseases, with existing drugs like abatacept and belatacept serving as precedents.[7]
A key element of ZB-004's design involves specific modifications within its CTLA-4 ECD component. The molecule incorporates two amino acid substitutions in this domain, although the precise identities of these substituted residues are not disclosed in the available abstracts.[3] The explicit purpose of these engineered changes to the CTLA-4 ECD is to augment the binding affinity of ZB-004 for its molecular targets: CD80 (also known as B7-1) and CD86 (also known as B7-2). These targets are crucial costimulatory molecules expressed on the surface of antigen-presenting cells (APCs), such as dendritic cells, macrophages, and B-cells.[3] Xencor's patent US9884902B2 details various mutations within the CTLA-4 ECD that can significantly enhance binding affinity to CD80 and CD86, providing a likely technological foundation for the specific modifications incorporated into ZB-004.[12]
The engineering of the CTLA-4 ECD for heightened affinity to CD80/CD86 represents a rational approach to potentially increase the therapeutic potency of ZB-004. An enhanced binding affinity could facilitate more effective blockade of the CD28-CD80/CD86 costimulatory axis, possibly at lower drug concentrations or achieving more complete and sustained target saturation, particularly in inflammatory microenvironments where ligand density might be high. The strength of the interaction between a therapeutic protein and its target is a direct determinant of its biological activity. Consequently, a higher affinity for CD80 and CD86 enables ZB-004 to more effectively outcompete the endogenous CD28 receptor for binding to these ligands.[3] This improved competitive binding could translate to a lower administered dose required to achieve the desired level of T-cell costimulation blockade, which in turn might improve the therapeutic index by minimizing potential off-target effects or dose-related toxicities.
The Fc domain of ZB-004 is a hybrid construct, incorporating elements from human IgG1 and IgG2 constant regions.[3] This hybrid design may be selected to optimize the balance between effector functions (or lack thereof, if ablated) and pharmacokinetic properties. A critical feature of this Fc domain is the inclusion of two specific amino acid substitutions: M428L (methionine at position 428 mutated to leucine) and N434S (asparagine at position 434 mutated to serine), according to EU numbering conventions. This modified Fc region is a component of Xencor's proprietary Xtend™ Fc technology platform.[3]
The Xtend™ Fc domain, by virtue of these M428L/N434S substitutions, is specifically engineered to exhibit increased binding affinity for the neonatal Fc receptor (FcRn).[3] FcRn is a pivotal receptor in maintaining IgG homeostasis. It binds to the Fc region of IgG within the acidic environment of endosomes following cellular pinocytosis. This binding rescues the IgG from lysosomal degradation and facilitates its recycling back to the cell surface, where it is released into the neutral pH of the bloodstream. An enhanced affinity for FcRn leads to more efficient recycling and a significantly reduced rate of catabolism, thereby prolonging the protein's persistence in circulation. Xencor's patent literature, such as US9884902B2 and US10793632B2, describes various Fc variants, including those with altered FcRn binding characteristics, designed to extend in vivo half-life.[12] The primary objective of incorporating this Fc engineering into ZB-004 is to achieve a marked extension of its in vivo circulating half-life, which would consequently allow for a reduced frequency of administration to patients.[3]
The successful application of Xtend™ Fc technology to ZB-004, as evidenced by the observed 8-18 day half-life in the Phase 1 study [3], represents a significant potential clinical and pharmacoeconomic advantage, particularly in the context of managing chronic autoimmune diseases. Conditions of this nature often necessitate long-term, if not lifelong, therapeutic intervention. A drug candidate with an extended half-life that permits substantially less frequent dosing regimens—for instance, subcutaneous injections administered every few weeks or even monthly, as opposed to more frequent schedules like weekly or bi-weekly—can dramatically improve patient convenience, reduce treatment burden, and foster better adherence to therapy. Enhanced adherence is a critical determinant of long-term treatment success. Moreover, from a healthcare system and patient perspective, a reduced dosing frequency can translate to lower annualized treatment costs, encompassing fewer drug administrations and potentially less frequent clinical monitoring. This attribute is a key differentiator for novel biologic agents entering competitive therapeutic landscapes.
ZB-004 exerts its immunomodulatory effects by specifically binding to the CD80 and CD86 molecules, which are expressed on the surface of APCs.[3] This binding action physically obstructs the interaction between CD80/CD86 on APCs and their cognate receptor, CD28, which is constitutively expressed on T-lymphocytes.[3]
The interaction between CD28 on T-cells and CD80/CD86 on APCs provides a critical "second signal," or costimulatory signal. This signal, in conjunction with the "first signal" delivered through the T-cell receptor (TCR) upon engagement with peptide-MHC (Major Histocompatibility Complex) complexes on APCs, is indispensable for the full activation, proliferation, cytokine production, and differentiation of T-cells into effector cells. By competitively inhibiting this vital CD28-mediated costimulatory pathway, ZB-004 effectively dampens T-cell-driven immune responses. This modulation is achieved without inducing broad immunosuppression or lymphocyte depletion, offering a more targeted approach. Such targeted immunomodulation is particularly relevant in the context of autoimmune diseases, where the dysregulation of autoreactive T-cells is a central driver of tissue damage and disease pathology.[7]
The mechanism of ZB-004, by focusing on the blockade of CD28-mediated costimulation, aims for a more refined immunomodulation compared to therapeutic strategies that involve widespread lymphocyte depletion or global inhibition of cytokine pathways. This specificity inherent in targeting "Signal 2" of T-cell activation may offer a more favorable balance between therapeutic efficacy and safety. Specifically, it might reduce the risk of opportunistic infections or other complications associated with broader immunosuppression, while still effectively controlling the pathogenic T-cell responses underlying autoimmune conditions. The advanced engineering of ZB-004, with its enhanced affinity and prolonged duration of action, is intended to optimize this targeted immunomodulatory effect, potentially leading to improved clinical outcomes for patients.
ZB-004 is being strategically developed for the treatment of a spectrum of autoimmune diseases, capitalizing on its mechanism of inhibiting T-cell costimulation.[1] The foundational development work was conducted by Xencor, Inc. Following the establishment of its preclinical profile, Zenas BioPharma acquired the rights to ZB-004 and has since been responsible for its clinical advancement.[2] Zenas BioPharma initiated Investigational New Drug (IND)-enabling studies [5] and subsequently progressed ZB-004 into Phase 1 clinical trials.[2] To date, the most advanced stage of clinical development completed for ZB-004 is Phase 1.[2]
The cornerstone of ZB-004's early clinical evaluation is the Phase 1 Single Ascending Dose (SAD) study conducted in healthy volunteers.
Table 2: Overview of Phase 1 SAD Clinical Trial (NCT05794516) Design
Feature | Details |
---|---|
Study Title | A Single Ascending Dose (SAD) Study to Evaluate the Safety and Pharmacokinetics (PK) of ZB004 in Healthy Volunteers |
NCT Number | NCT05794516 |
Phase | Phase 1 |
Sponsor | Zenas BioPharma LLC |
Design | Randomized, double-blind, placebo-controlled, single ascending dose |
Population | Healthy Volunteers (N=40 total; 5 cohorts of 8 participants each, 3:1 active:placebo randomization per cohort), ages 18-55 years |
Intervention | ZB-004, single subcutaneous (SC) injection |
Dose Levels | 3 mg, 12.5 mg, 50 mg, 125 mg, 200 mg |
Control | Placebo, single subcutaneous (SC) injection |
Location | New Zealand Clinical Research (single center) |
Status | Completed |
Primary Outcome Measures | Safety and Tolerability: Incidence of treatment-emergent adverse events (TEAEs), serious adverse events (SAEs) |
Secondary Outcome Measures | Pharmacokinetics (PK): Cmax, Tmax, AUCinf, AUClast, half-life<br>Immunogenicity: Anti-drug antibody (ADA) incidence and titer<br>Pharmacodynamics (PD): CD86 receptor occupancy, ex vivo IL-2 inhibition |
This table consolidates the critical design elements of the first-in-human study for ZB-004, offering a structured and accessible overview of the trial's methodology and objectives. This framework is essential for interpreting the subsequent results and appreciating the scientific rigor of ZB-004's early clinical evaluation. The information is derived from multiple consistent sources.[2]
The results from the Phase 1 single ascending dose study of ZB-004 in healthy volunteers have provided crucial initial data on its pharmacokinetic profile, pharmacodynamic activity, safety, and immunogenicity.[3]
The pharmacokinetic evaluation of ZB-004 yielded several key findings:
Table 3: Summary of Key Pharmacokinetic (PK) Parameters from NCT05794516
Parameter | Reported Value/Observation |
---|---|
Terminal Half-life (t1/2) | 8 – 18 days |
Dose Proportionality (Cmax, AUC) | Non-linear increase with increasing dose |
Target-Mediated Drug Disposition (TMDD) | Evidence of TMDD effect observed |
This table succinctly presents the most critical pharmacokinetic findings for ZB-004 from its initial human study. The extended half-life is a key attribute supporting the potential for less frequent dosing, a significant advantage for chronic therapies. The observation of TMDD is pharmacologically important as it confirms potent target binding in vivo, a prerequisite for achieving the desired therapeutic effect. These data are crucial for designing subsequent multiple-dose studies and for understanding the dose-exposure-response relationships of ZB-004.[3]
The pharmacodynamic assessments provided direct evidence of ZB-004's biological activity and target engagement in healthy volunteers:
The pharmacodynamic data from this Phase 1 study are particularly significant. They establish a clear link between the administration of ZB-004, its engagement with the cell-surface target CD86 in a dose-responsive manner, and a consequent functional immunomodulatory effect (inhibition of IL-2 production). CD86 is a crucial ligand for the CD28 costimulatory receptor on T-cells. By occupying CD86, ZB-004 effectively blocks the CD28-mediated costimulatory signal necessary for full T-cell activation, which in turn leads to reduced IL-2 synthesis. This demonstration of a clear pharmacokinetic/pharmacodynamic (PK/PD) relationship in an early-phase human study provides strong validation for the drug's intended mechanism of action and is instrumental in guiding dose selection for subsequent therapeutic trials in patient populations.
Table 4: Summary of Key Pharmacodynamic (PD) Markers from NCT05794516
PD Marker | Key Finding |
---|---|
CD86 Receptor Occupancy (RO) in whole blood | Clear target engagement; dose-dependent increase in %CD86 RO. |
Ex vivo stimulated IL-2 production | Inhibition of IL-2 production observed in all dose cohorts. |
This table highlights that ZB-004 is not merely present in the systemic circulation (as shown by PK data) but is also biologically active at its intended molecular target. The ability to demonstrate both target engagement and a relevant downstream functional effect in a Phase 1 study is a critical milestone. It significantly de-risks further development by providing confidence in the drug's proposed mechanism of action and its potential to effectively modulate immune responses in individuals with autoimmune diseases.[3]
The evaluation of safety and tolerability was a primary objective of the NCT05794516 study.
Table 5: Summary of Safety and Tolerability Findings from NCT05794516
Safety Aspect | Finding |
---|---|
Overall Tolerability | Well-tolerated at all single SC doses studied (up to 200 mg). |
Adverse Events (AEs) | Mild; consistent with expectations in healthy volunteers; no new signals. |
Serious Adverse Events (SAEs) | None observed. |
Key Safety Conclusion | Favorable safety and tolerability profile in healthy volunteers. |
Establishing a robust safety and tolerability profile in Phase 1 is a fundamental prerequisite for the continued development of any investigational therapeutic. The findings for ZB-004, as summarized in this table, are positive, indicating that the drug, at the single doses tested, did not elicit undue safety concerns in healthy individuals. This favorable outcome is critical for supporting the progression of ZB-004 into studies involving patient populations with autoimmune diseases, where the risk-benefit assessment will be further refined.[3]
The potential for ZB-004 to elicit an immune response in the form of anti-drug antibodies (ADAs) was assessed as a secondary objective.
The observation that higher doses of an immunomodulatory agent like ZB-004 are associated with a reduction in its own immunogenicity is a significant and highly favorable finding. Typically, increased exposure to a therapeutic protein might be anticipated to elevate the risk of an immune response against it. However, in the case of ZB-004, its primary pharmacological activity—the inhibition of T-cell costimulation—is likely dampening the T-cell dependent immune response that would be necessary for the generation of high-affinity ADAs against the ZB-004 protein itself. As higher doses of ZB-004 achieve greater systemic immunosuppression, this effect appears to extend to mitigating the immune system's ability to recognize and mount a response to ZB-004 as a foreign antigen. This characteristic is particularly desirable for a biologic therapeutic intended for chronic administration in the context of autoimmune diseases. A reduced risk of developing clinically relevant ADAs could translate to more sustained efficacy over time, a lower likelihood of altered pharmacokinetic profiles due to ADA binding, and a diminished potential for ADA-mediated hypersensitivity reactions or other adverse events. This favorable immunogenicity trend, if maintained in patient populations, would represent a substantial advantage for ZB-004.
The development of ZB-004 is underpinned by a robust intellectual property (IP) framework, primarily stemming from Xencor's proprietary XmAb® antibody engineering technologies.
The multi-faceted bioengineering strategy employed in ZB-004, which integrates proprietary modifications to both the ligand-binding CTLA-4 ECD and the pharmacokinetic-modulating Fc domain (Xtend™ technology), results in a highly differentiated molecular entity. This intricate design, combining enhanced target affinity with an extended half-life, is likely protected by a layered and robust intellectual property portfolio. Xencor's established expertise and comprehensive patent estate in the field of Fc engineering and antibody optimization [11] form a strong foundation for this IP position. Zenas BioPharma, through its licensing agreement, leverages this existing IP to secure exclusive rights for the continued development and commercialization of ZB-004. This strong IP protection is crucial for attracting investment, supporting long-term development efforts, and ultimately, ensuring market exclusivity for ZB-004 should it achieve regulatory approval.
The Phase 1 single ascending dose study (NCT05794516) of ZB-004 in healthy volunteers successfully met its predefined objectives. The data clearly demonstrated that ZB-004 is safe and well-tolerated at subcutaneous doses up to 200 mg.[3] The pharmacokinetic analysis confirmed the attainment of an extended plasma half-life, ranging from 8 to 18 days, which aligns with the intended design benefit of the Xtend™ Fc technology and supports the potential for less frequent dosing regimens in a clinical setting. Furthermore, the observation of target-mediated drug disposition (TMDD) provided in vivo evidence of potent and specific binding of ZB-004 to its targets, CD80 and CD86.[3]
Pharmacodynamically, the study yielded compelling results. Clear, dose-dependent target engagement was demonstrated through measurements of CD86 receptor occupancy. This was complemented by evidence of functional immunomodulation, as indicated by the inhibition of ex vivo stimulated IL-2 production across all dose cohorts.[3] These PD findings confirm that ZB-004 not only reaches and binds its target but also elicits a biological response consistent with its proposed mechanism of T-cell costimulation blockade. The immunogenicity profile observed was also favorable, with an intriguing trend towards lower incidence and titers of anti-drug antibodies (ADAs) at higher doses of ZB-004, potentially attributable to the drug's own immunosuppressive effects.[3]
The bioengineered attributes of ZB-004—enhanced affinity for CD80/CD86 and an extended circulating half-life—carry significant implications for its potential clinical utility in autoimmune diseases:
The combination of these engineered improvements—potentially higher potency stemming from enhanced target affinity and a markedly improved dosing schedule enabled by the extended half-life—positions ZB-004 as a potentially differentiated, next-generation CTLA-4-Ig therapeutic. In the competitive landscape of autoimmune disease treatments, new entrants must offer clear and tangible advantages over existing standards of care. Abatacept, the first-generation CTLA-4-Ig, has established the clinical utility of this mechanism but typically requires weekly subcutaneous injections or monthly intravenous infusions. A therapy like ZB-004, if its efficacy and safety are confirmed in patient populations, could offer a similar or potentially improved therapeutic effect with a much more patient-friendly and convenient dosing regimen. This dual advantage of potentially superior pharmacology and enhanced convenience could make ZB-004 a preferred therapeutic option for both patients and prescribing clinicians in the future.
The fundamental mechanism of T-cell costimulation blockade via CTLA-4-Ig fusion proteins is a clinically validated therapeutic strategy for a range of T-cell-mediated autoimmune diseases. Conditions such as rheumatoid arthritis, psoriatic arthritis, and juvenile idiopathic arthritis have seen significant treatment advances with the use of abatacept.[7] The broad applicability of this mechanism suggests that ZB-004 could also have therapeutic potential across a similar spectrum of autoimmune disorders.
Zenas BioPharma, the current developer of ZB-004, has a clear strategic focus on immunology and the development of therapies for autoimmune diseases. This is evident from its broader pipeline, which includes other immunomodulatory candidates such as obexelimab (a bifunctional antibody targeting CD19 and FcγRIIb) and ZB002 (an anti-TNFα therapy).[2] While the specific autoimmune indications that Zenas BioPharma will prioritize for ZB-004 development beyond the general classification of "Autoimmune Diseases" [2] are not explicitly detailed in the currently available information, the company's overall focus and the drug's mechanism of action suggest a wide range of potential applications. The selection of lead indications for Phase 2 and Phase 3 development will likely be a strategic decision informed by several factors, including the strength of the scientific rationale for CTLA-4 pathway involvement in specific diseases, the magnitude of unmet medical need, the existing standard of care and competitive landscape in those indications, and an assessment of where ZB-004's potentially differentiated profile (e.g., less frequent subcutaneous dosing, potentially higher potency) might offer the most compelling clinical benefit and market opportunity.
The positive outcomes from the Phase 1 SAD study in healthy volunteers provide a strong rationale for advancing ZB-004 into further clinical development, specifically into Phase 2 proof-of-concept studies in patient populations with selected autoimmune diseases. Several key considerations will shape this future development:
While the robust Phase 1 data have significantly de-risked the early development of ZB-004 from the perspectives of safety, pharmacokinetics, and confirmation of its mechanism of action, the path to demonstrating definitive clinical efficacy in specific autoimmune diseases remains a challenging endeavor. Autoimmune diseases are often complex and heterogeneous, and patient responses to immunomodulatory therapies can vary considerably. Translating the favorable profile observed in healthy volunteers into clinically meaningful improvements in disease activity, patient-reported outcomes, and overall quality of life in specific patient populations will be the next major developmental hurdle. The competitive environment within the autoimmune therapeutic space is intense, with numerous existing and emerging therapies. The ultimate success of ZB-004 will hinge on its ability to demonstrate a clear and compelling clinical advantage—whether in terms of superior efficacy, an improved safety profile, or enhanced patient convenience and adherence—in well-chosen indications. Nevertheless, the strong foundational science, meticulous bioengineering, and positive early clinical data for ZB-004 provide a promising basis for its continued development by Zenas BioPharma.
ZB-004 (XmAb10717) is a rationally designed, second-generation CTLA-4-Ig fusion protein that incorporates advanced bioengineering features aimed at enhancing its therapeutic properties for the treatment of autoimmune diseases. Key among these features are modifications to the CTLA-4 extracellular domain for increased binding affinity to its targets (CD80/CD86) and the incorporation of Xencor's Xtend™ Fc technology (M428L/N434S substitutions) to achieve a significantly extended pharmacokinetic half-life.
The recently completed Phase 1 single ascending dose clinical trial (NCT05794516) in healthy volunteers has provided critical initial validation of ZB-004's profile. The study demonstrated that ZB-004 is safe and well-tolerated at subcutaneous doses up to 200 mg. Pharmacokinetic analyses confirmed the intended extended half-life of 8 to 18 days, supporting the potential for less frequent dosing regimens, and revealed target-mediated drug disposition indicative of potent in vivo target binding. Furthermore, pharmacodynamic assessments provided clear evidence of dose-dependent target engagement, as measured by CD86 receptor occupancy, and functional immunomodulation, demonstrated by the inhibition of ex vivo stimulated IL-2 production. The immunogenicity profile of ZB-004 also appears favorable, with an observed trend towards lower anti-drug antibody formation at higher doses, possibly due to the drug's inherent immunosuppressive activity.
Collectively, these Phase 1 findings are highly encouraging and provide a strong foundation for the continued clinical development of ZB-004 by Zenas BioPharma. ZB-004 stands as a promising therapeutic candidate with the potential to offer an improved clinical profile, including enhanced patient convenience, compared to existing therapies for a range of autoimmune diseases. Future clinical studies in patient populations will be essential to definitively establish its efficacy and long-term safety in specific autoimmune indications.
Published at: May 13, 2025
This report is continuously updated as new research emerges.