ZW-191 is an investigational antibody-drug conjugate (ADC) engineered by Zymeworks Inc., targeting Folate Receptor alpha (FRα), a validated cell-surface antigen overexpressed in various solid tumors with high unmet medical need. The molecular architecture of ZW-191 comprises a novel humanized IgG1 antibody, a proprietary potent topoisomerase I inhibitor (TOPO1i) payload designated ZD06519, and a protease-cleavable linker system (maleimidocaproyl anchor with a GGFG-aminomethyl sequence), achieving a drug-to-antibody ratio (DAR) of 8.[1] This design reflects a strategic approach to optimize therapeutic efficacy, particularly in tumor microenvironments characterized by heterogeneous antigen expression, leveraging superior antibody internalization and a potent bystander-active payload.
Preclinical evaluations have demonstrated ZW-191's potent and selective anti-tumor activity. In vitro, the ADC exhibited sub-nanomolar binding affinity to FRα, superior cellular internalization, and efficient payload delivery, leading to cytotoxicity in FRα-expressing cancer cell lines and 3D tumor spheroids.[1] A significant bystander effect, mediated by the cell-permeable ZD06519 payload, was observed, enabling the killing of adjacent FRα-negative tumor cells.[1] In vivo, ZW-191 induced substantial tumor regressions in patient-derived xenograft (PDX) models of ovarian cancer, non-small cell lung cancer (NSCLC), endometrial cancer, and triple-negative breast cancer (TNBC), critically, across a spectrum of FRα expression levels (high, mid, and low).[1] Comparative studies indicated superior or comparable activity to mirvetuximab soravtansine, especially in models with lower FRα expression.[2]
Non-human primate (NHP) toxicology studies revealed an encouraging safety profile, with ZW-191 being well-tolerated up to 60 mg/kg (Q3Wx3), established as the highest non-severely toxic dose (HNSTD).[2] Observed effects, primarily gastrointestinal and transient hematologic/biochemical changes, were generally non-adverse and reversible, and notably, no ophthalmic toxicities were reported.[2] The pharmacokinetic profile in NHPs was deemed favorable for clinical development.[1]
Clinically, ZW-191 is advancing under an Investigational New Drug (IND) application cleared by the U.S. Food and Drug Administration (FDA) in July 2024.[5] A global, multi-center, open-label Phase 1 clinical trial (NCT06555744; ZWI-ZW191-101) was initiated, with the first patient dosed in November 2024.[7] This two-part study aims to evaluate the safety, tolerability, pharmacokinetics, and preliminary anti-tumor activity of ZW-191 in approximately 145 adult patients with advanced FRα-expressing solid tumors, including ovarian, endometrial, and NSCLC.[10] The development of ZW-191, particularly its potential efficacy in tumors with low or heterogeneous FRα expression, positions it to address significant unmet medical needs and could validate Zymeworks' broader ADC platform technologies.
Antibody-drug conjugates (ADCs) have emerged as a transformative class of therapeutics in oncology, designed to selectively deliver potent cytotoxic agents to cancer cells while minimizing systemic exposure and associated toxicities. This targeted approach is achieved by linking a monoclonal antibody, specific for a tumor-associated antigen, to a cytotoxic payload via a stable linker. The evolving landscape of ADC technology focuses on optimizing each component—antibody, linker, and payload—to enhance the therapeutic index.
Within this context, Zymeworks Inc. is developing ZW-191, a novel investigational ADC targeting Folate Receptor alpha (FRα).[1] FRα is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein with limited expression in normal adult tissues but is frequently overexpressed in a variety of epithelial malignancies. These include ovarian cancer, endometrial cancer, non-small cell lung cancer (NSCLC), and triple-negative breast cancer (TNBC), among others, making it a clinically validated target for ADC development.[1] The therapeutic rationale for targeting FRα is underscored by the clinical validation provided by mirvetuximab soravtansine, an FRα-targeting ADC approved for certain ovarian cancers.[2]
Despite the clinical validation of FRα as an ADC target, a significant unmet need persists, particularly for patients whose tumors exhibit low or heterogeneous FRα expression. For instance, it is estimated that approximately two-thirds of ovarian cancer patients may not be eligible for treatment with mirvetuximab soravtansine due to insufficient FRα expression levels based on current diagnostic criteria.[2] ZW-191 has been engineered with distinct molecular features, including a novel antibody with enhanced internalization properties and a potent bystander-active payload, aimed at addressing these limitations. Preclinical data suggest that ZW-191 may offer improved efficacy, especially in tumors with lower FRα expression, potentially expanding the patient population that could benefit from FRα-targeted ADC therapy.[2] The development of ZW-191 thus represents an effort to build upon existing knowledge in FRα targeting, aiming to deliver a more effective therapeutic option across a broader range of FRα-expressing cancers.
ZW-191 is a complex macromolecular therapeutic, meticulously designed with distinct components that synergistically contribute to its targeted anti-tumor activity. Its structure and mechanism reflect advancements in ADC engineering aimed at maximizing efficacy and improving the therapeutic window.
Table 1: Summary of ZW-191 Molecular Characteristics
Component | Description/Properties | Source Snippet(s) |
---|---|---|
Antibody | Novel, fully humanized IgG1; targets human Folate Receptor alpha (FRα); engineered for superior internalization, payload delivery, and tumor spheroid penetration; sub-nanomolar binding affinity. | 1 |
Payload | ZD06519: a novel, proprietary, rationally designed, moderate potency, bystander-active camptothecin-based topoisomerase I inhibitor (TOPO1i). | 1 |
Linker | Maleimidocaproyl (MC) anchor and a glycyl glycyl phenylalanyl glycyl (GGFG)-aminomethyl (AM) protease-cleavable sequence; conjugates payload to antibody via endogenous interchain cysteines. | 1 |
DAR | Drug-to-Antibody Ratio of 8; selected to balance efficacy and tolerability. | 2 |
Antibody Component:
The targeting moiety of ZW-191 is a novel, fully humanized immunoglobulin G1 (IgG1) monoclonal antibody.2 This antibody was specifically selected for its high affinity and specificity for human FRα, exhibiting sub-nanomolar binding capabilities.2 A critical feature of this antibody is its enhanced internalization characteristics. Compared to other FRα-targeted antibodies used in ADC development, the ZW-191 antibody has demonstrated superior efficiency in cellular internalization following FRα binding, leading to more effective payload delivery into the tumor cell and enhanced penetration into tumor spheroids, which mimic aspects of the solid tumor microenvironment.1 This improved intracellular trafficking is a key attribute, as efficient delivery of the cytotoxic payload to its site of action is paramount for ADC efficacy.
Payload: ZD06519
The cytotoxic component of ZW-191 is ZD06519, a novel, proprietary topoisomerase I inhibitor (TOPO1i) based on a camptothecin scaffold.1 ZD06519 was rationally designed for moderate potency and, significantly, possesses bystander activity.2 Topoisomerase I is an essential enzyme involved in DNA replication and transcription; its inhibition leads to DNA strand breaks and ultimately, apoptotic cell death. The bystander effect means that once ZD06519 is released within a target FRα-positive cell, it can diffuse through cell membranes to kill nearby FRα-negative tumor cells, a property particularly advantageous in tumors with heterogeneous antigen expression.1 Zymeworks has highlighted ZD06519 as a key component of its ADC platform, with plans to utilize this payload in other ADC candidates (ZW220 and ZW251).5
Linker Technology:
ZW-191 employs a protease-cleavable linker system to attach the ZD06519 payload to the antibody. This linker consists of a maleimidocaproyl (MC) anchor, which facilitates conjugation to cysteine residues on the antibody, and a peptide sequence, glycyl glycyl phenylalanyl glycyl (GGFG)-aminomethyl (AM).1 The GGFG motif is designed to be selectively cleaved by lysosomal proteases, such as cathepsin B, which are abundant within the intracellular environment of tumor cells after ADC internalization. This ensures that the payload is preferentially released within the target cell, minimizing premature release in systemic circulation and thereby reducing off-target toxicity. The conjugation occurs via endogenous interchain cysteines of the antibody.2
Drug-to-Antibody Ratio (DAR):
ZW-191 is engineered with a drug-to-antibody ratio (DAR) of 8.2 This relatively high DAR means that, on average, eight molecules of the ZD06519 payload are attached to each antibody molecule. The selection of DAR 8 was a deliberate choice aimed at maximizing the cytotoxic payload delivered per antibody binding event, while seeking an optimal balance between anti-tumor efficacy and systemic tolerability.5
Detailed Mechanism of Action (MOA):
The anti-tumor activity of ZW-191 is a multi-step process:
The combination of a highly internalizing antibody, a potent TOPO1i payload with bystander activity, a stable yet cleavable linker, and a high DAR positions ZW-191 as an ADC engineered to overcome several common challenges in ADC therapy, such as poor tumor penetration and efficacy against tumors with heterogeneous or low antigen expression. This molecular design underpins its potential for a broad therapeutic window and activity across multiple cancer types.
The preclinical development of ZW-191 involved extensive in vitro and in vivo studies to characterize its pharmacological properties and anti-tumor efficacy. These studies have provided a strong rationale for its advancement into clinical trials.
In Vitro Studies:
In Vivo Anti-Tumor Activity (Patient-Derived Xenograft - PDX models):
ZW-191's anti-tumor activity was evaluated in a diverse panel of PDX models, which are generally considered more representative of human tumor biology than cell line-derived xenografts. Tumor regression was defined as a tumor volume change of less than 0% from baseline.2
Table 2: Illustrative Preclinical Efficacy Data of ZW-191 in Selected PDX Models
PDX Model | Cancer Type | FRα Expression (H-Score) | ZW-191 Dose (mg/kg, single) | ZW-191 Incidence of Tumor Regression | Mirvetuximab Soravtansine Dose (mg/kg, single) | Mirvetuximab Soravtansine Incidence of Tumor Regression | Source Snippet(s) |
---|---|---|---|---|---|---|---|
OVXF_630_T | Ovarian Cancer | 240 | 6 | 3/3 | 6 | 3/3 | 2 |
OVXF_1320_T | Ovarian Cancer | 30 | 6 | 3/3 | 6 | 0/3 | 2 |
LGXF_2121_T | NSCLC | 210 | 6 | Not explicitly stated, strong TGI | 6 | Not explicitly stated, less TGI | 2 |
LANOXF_A002_A | NSCLC | 80 | 6 | Tumor Regression (visual) | 6 | No Regression (visual) | 2 |
ENDOXF_A001_B | Endometrial Cancer | 165 | 6 | Tumor Regression (visual) | 6 | Less Regression (visual) | 2 |
BRXF_2553_T | TNBC | 80 | 6 | Tumor Regression (visual) | 6 | No Regression (visual) | 2 |
Note: "Tumor Regression" typically defined as <0% change from baseline tumor volume. TGI = Tumor Growth Inhibition. H-score determined by pathologist from research level IHC assay. Data often presented visually in source figures.
The consistent outperformance or equivalence of ZW-191 against mirvetuximab soravtansine, particularly in models with low FRα expression, supports the hypothesis that ZW-191's unique design attributes—specifically its highly internalizing antibody and potent bystander payload—may translate to clinical benefit in a broader patient population than currently addressable by existing FRα-targeted ADCs. The robust bystander effect is a critical component of this potential, offering a mechanism to overcome intra-tumoral heterogeneity, a common cause of therapeutic resistance.
To assess the safety and pharmacokinetic profile of ZW-191 prior to human clinical trials, comprehensive studies were conducted in non-human primates (NHPs), specifically cynomolgus monkeys. These studies are crucial for identifying potential toxicities, establishing a safe starting dose for human trials, and understanding the drug's behavior in a biological system more complex than rodent models.
Pharmacokinetic (PK) Profile:
ZW-191 demonstrated a favorable pharmacokinetic profile in NHPs.1 While specific PK parameters such as half-life, clearance, and area under the curve (AUC) were not detailed in the available abstracts, the overall assessment indicated that the PK characteristics support the planned clinical dosing schedules, typically involving intermittent administration (e.g., every three weeks, Q3W).2
Safety and Tolerability Studies:
A Good Laboratory Practice (GLP) repeat-dose toxicology study was performed in NHPs, with ZW-191 administered every three weeks for three cycles (Q3Wx3).2
Dose Level (mg/kg) | Key Clinical Observations | Histopathological Findings | Clinical Chemistry Changes | Overall Assessment (Adverse/Non-Adverse, Reversibility) | Source Snippet(s) |
---|---|---|---|---|---|
10 | No adverse effects reported | No adverse effects reported | Slight, transient ↑ AST, ALT (n=1) | Non-adverse, Reversible | 2 |
30 | Emesis/vomitus | ↓ Thymic lymphocytes, ↓ Pancreatic acinar cell secretion | ↑ AST, ALT | Non-adverse, Reversible | 2 |
60 | Liquid/discolored feces, Emesis/vomitus, ↓ Activity level (n=1) | ↓ Thymic lymphocytes, ↓ Pancreatic acinar cell secretion | ↑ AST, ALT, ↑ Creatine Kinase (CK) | Non-adverse, Reversible | 2 |
*AST = Aspartate Aminotransferase; ALT = Alanine Aminotransferase; CK = Creatine Kinase.*
The NHP toxicology data, particularly the establishment of an HNSTD at 60 mg/kg and the reversible nature of the observed effects (primarily gastrointestinal disturbances and transient changes in lymphocyte counts, pancreatic enzyme indicators, and liver enzymes), provided a solid basis for proceeding to Phase 1 clinical trials. The absence of irreversible or life-threatening toxicities at clinically relevant exposures was encouraging. However, the consistent findings of decreased thymic lymphocytes and pancreatic acinar cell secretion at the higher doses (30 and 60 mg/kg), even if deemed non-adverse and reversible in NHPs, highlight these as areas for careful monitoring in human subjects during early clinical development. The clinical significance of these observations, especially under conditions of repeated dosing in patients, will need to be thoroughly evaluated.
Following the promising preclinical data and favorable NHP toxicology profile, ZW-191 has transitioned into clinical development.
Regulatory Milestone:
The Investigational New Drug (IND) application for ZW-191 was cleared by the U.S. Food and Drug Administration (FDA) in July 2024.5 This clearance paved the way for the initiation of the first-in-human clinical trial. Zymeworks also indicated plans to file for regulatory authorization to commence clinical studies in non-US jurisdictions in the second half of 2024.5
Study Identification:
The ongoing Phase 1 clinical trial is identified as:
Study Design:
The ZWI-ZW191-101 study is a Phase 1, first-in-human, open-label, multi-center, global trial.7 It is structured in two parts:
Table 4: Overview of NCT06555744 (ZWI-ZW191-101) Clinical Trial Design
Feature | Details | Source Snippet(s) |
---|---|---|
Study Title | A Study of ZW191 in Participants With Solid Tumors | 10 |
ClinicalTrials.gov ID | NCT06555744 | 7 |
Phase | Phase 1 | 3 |
Study Design | Two-part (Dose Escalation, Dose Expansion), Open-label, Multi-center, Global, Single-arm (in dose escalation) | 10 |
Primary Objectives | Part 1: Evaluate safety and tolerability of ZW-191, determine MTD/RP2D. Part 2: Further evaluate safety and explore potential anti-tumor activity. | 7 |
Key Secondary Objectives | Assess pharmacokinetics (PK) and confirmed objective response rate (ORR) per RECIST v1.1. | 7 |
Target Indications | Advanced FRα-expressing solid tumors, specifically including ovarian cancer, endometrial cancer, and non-small cell lung cancer (NSCLC). | 3 |
Key Inclusion Criteria | Pathologically/cytologically confirmed advanced (unresectable), recurrent/metastatic disease; measurable disease per RECIST v1.1; ECOG PS 0 or 1; LVEF ≥ 50%; adequate organ function. | 10 |
Key Exclusion Criteria | Progressing additional malignancy requiring active treatment; prior Topoisomerase I inhibitor (TOPO1i) ADC treatment; uncontrolled renal/pancreatic/liver disease; severe active infections. | 10 |
Estimated Enrollment | Approximately 145 adult patients. | 7 |
Current Status | Active, Recruiting. | 7 |
Key Timelines | IND Cleared: July 2024. First Patient Dosed: November 2024. Planned TiP Poster: ESMO Gynaecological Cancers Congress, June 2025. | 5 |
Target Population:
The study aims to enroll approximately 145 adult patients with pathologically or cytologically confirmed advanced FRα-expressing solid tumors that are locally advanced (unresectable), recurrent, or metastatic.7 Specific tumor types of interest include ovarian cancer, endometrial cancer, and NSCLC.3
Key inclusion criteria mandate measurable disease according to RECIST v1.1, an Eastern Cooperative Oncology Group (ECOG) Performance Status score of 0 or 1, adequate cardiac function (defined as Left Ventricular Ejection Fraction (LVEF) ≥ 50%), and other adequate organ function.10
Notable exclusion criteria include any known additional malignancy that is progressing or requires active treatment, or that may interfere with study endpoints. Critically, patients who have received prior treatment with a Topoisomerase I inhibitor (TOPO1i) ADC are excluded, regardless of the washout period.10 Other exclusions comprise acute or chronic uncontrolled renal disease, pancreatitis, or liver disease, and severe chronic or active infections requiring systemic therapy.10 The exclusion of patients with prior TOPO1i ADC exposure is a significant aspect of the trial design. This ensures that the initial assessment of ZW-191's safety and efficacy is not confounded by potential pre-existing resistance mechanisms or cumulative toxicities from similar payloads. While this approach provides a cleaner dataset for initial evaluation, it means that the trial will not immediately address the activity of ZW-191 in the TOPO1i-ADC refractory setting, which is an increasingly relevant clinical scenario as TOPO1i ADCs become more widely used.
Current Status and Timelines:
The ZWI-ZW191-101 trial is currently active and recruiting participants.7 The first patient was dosed in November 2024.7 The relatively rapid transition from IND clearance in July 2024 to the first patient being dosed suggests efficient operational planning and a high priority for the ZW-191 program within Zymeworks.
Trial Locations:
The study is being conducted at investigator sites across North America (including Connecticut and Ohio in the US), Europe, and the Asia-Pacific region.7 The NCI website lists three active locations.20 The global nature of the trial is intended to facilitate timely patient accrual and generate data applicable to diverse regulatory environments.
Planned Communications:
Zymeworks plans to present a Trial-in-Progress (TiP) poster for the ZWI-ZW191-101 study at the European Society of Medical Oncology (ESMO) Gynaecological Cancers Congress, scheduled for June 19-21, 2025.14 This will likely be one of the first public disclosures of the trial design and early progress to the oncology community.
The development of ZW-191 is grounded in a strong therapeutic rationale centered on the characteristics of its target, FRα, and the specific design features of the ADC intended to address unmet clinical needs.
Folate Receptor Alpha (FRα) as a Therapeutic Target:
Addressing Unmet Needs:
ZW-191 aims to address several unmet needs in the treatment of FRα-expressing cancers:
Potential Advantages of ZW-191:
Based on its design and preclinical data, ZW-191 may offer several advantages:
The successful clinical development of ZW-191, particularly if it demonstrates efficacy in tumors with low or heterogeneous FRα expression, could not only provide a new treatment option for patients but also stimulate further research into optimizing ADC components for targets that present similar expression challenges. This could influence broader strategies in ADC development for various solid tumors.
The regulatory journey and future development path for ZW-191 are critical determinants of its potential translation into a clinical therapy.
Current Regulatory Milestones:
Special Designations:
Based on the provided information, ZW-191 has not yet received any specific FDA or European Medicines Agency (EMA) special designations such as Orphan Drug Designation, Fast Track Designation, or Breakthrough Therapy Designation.14 While Zymeworks has experience securing such designations for other product candidates in its pipeline (e.g., zanidatamab received Fast Track and Breakthrough Therapy designations 29), these have not been reported for ZW-191 to date. The absence of these designations at this early stage is not unusual, as they are often granted based on emerging clinical data that demonstrates a potential to address serious conditions and unmet medical needs, or offer substantial improvement over available therapies. As clinical data from the Phase 1 trial become available, Zymeworks may pursue such designations if the results are compelling.
Future Perspectives:
The future development of ZW-191 hinges on the outcomes of its ongoing clinical program:
ZW-191's development timeline, with IND clearance in mid-2024 and the first patient dosed by late 2024, positions it within a competitive and rapidly evolving field of FRα-targeting ADCs and TOPO1i-based ADCs. Its ability to quickly generate compelling Phase 1 data demonstrating a favorable safety profile and efficacy, especially in tumors with low or heterogeneous FRα expression, will be crucial for its differentiation and continued advancement.
ZW-191 emerges from preclinical studies as a rationally designed, next-generation antibody-drug conjugate with several features that suggest potential advantages in the treatment of Folate Receptor alpha (FRα)-expressing solid tumors. Its core design elements—a novel humanized IgG1 antibody with superior internalization properties, a potent and bystander-active topoisomerase I inhibitor payload (ZD06519), a stable yet protease-cleavable linker, and a high drug-to-antibody ratio of 8—collectively aim to maximize targeted payload delivery and anti-tumor efficacy, particularly in challenging tumor contexts.[1]
The preclinical data package for ZW-191 is compelling. The consistent demonstration of potent in vitro cytotoxicity, effective bystander killing, and significant in vivo anti-tumor activity in diverse patient-derived xenograft models—importantly, including those with low and heterogeneous FRα expression—highlights its potential to address a broader patient population than some existing FRα-targeted therapies.[1] The head-to-head preclinical superiority or comparability to mirvetuximab soravtansine, especially in low FRα settings, further underscores this potential.[2] The non-human primate toxicology studies indicate a manageable safety profile, with an HNSTD of 60 mg/kg and reversible, non-adverse findings at higher doses, and notably, an absence of ophthalmic toxicity, which can be a concern with other ADC classes.[2]
The program has smoothly transitioned into early-stage clinical development with the FDA clearance of its IND in July 2024 and the initiation of the global Phase 1 trial (NCT06555744; ZWI-ZW191-101) in November 2024.[5] This trial will provide the first human data on the safety, tolerability, pharmacokinetics, and preliminary efficacy of ZW-191 in patients with advanced FRα-expressing cancers, including ovarian, endometrial, and non-small cell lung cancers.
Several factors will be critical to ZW-191's future success. Firstly, the translation of the NHP safety profile to humans will be closely watched. While the observed toxicities in NHPs (such as decreased thymic lymphocytes and pancreatic acinar cell secretion) were deemed non-adverse and reversible, their clinical manifestation and manageability in cancer patients, who may have different underlying physiological states and co-morbidities, will need careful evaluation.[2] Secondly, managing potential TOPO1i class-related toxicities (e.g., myelosuppression, gastrointestinal effects) will be important, although the specific profile of ZD06519 may differ from other TOPO1i payloads. Thirdly, the competitive landscape for FRα-targeted therapies and for ADCs employing TOPO1i payloads is dynamic and expanding. ZW-191's differentiation, particularly its efficacy in low FRα expressors and its bystander mechanism, will need to be clearly demonstrated in clinical settings to establish its unique value proposition.
The upcoming Trial-in-Progress poster at the ESMO Gynaecological Cancers Congress in June 2025 is anticipated to provide early insights into the clinical trial's conduct.[14] Subsequent data readouts from the Phase 1 study will be pivotal in validating the therapeutic potential and safety of ZW-191. If successful, ZW-191 could not only offer a significant new treatment option for patients with a range of FRα-expressing cancers, potentially including those currently underserved by existing therapies, but also validate Zymeworks' ZD06519 payload technology for its broader ADC pipeline. The journey of ZW-191 from preclinical promise to potential clinical impact is at an early but critical stage, with the oncology community keenly awaiting the emergence of human data.
Published at: May 28, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.