2169946-15-8
Zanidatamab represents a significant advancement in the field of targeted oncology, establishing a new class of therapy as a biparatopic, human epidermal growth factor receptor 2 (HER2)-directed antibody. Marketed under the brand name Ziihera®, this engineered biologic possesses a novel mechanism of action that distinguishes it from prior generations of HER2-targeted agents. By simultaneously engaging two distinct, non-overlapping epitopes on the HER2 receptor, zanidatamab induces unique receptor clustering, leading to rapid internalization, downregulation, and a multi-pronged, immune-mediated attack on tumor cells that includes potent complement-dependent cytotoxicity (CDC)—a mechanism largely absent in its predecessors.
The clinical value of this unique pharmacology was definitively established in the pivotal HERIZON-BTC-01 trial, a global Phase 2b study in patients with previously treated, unresectable or metastatic HER2-positive biliary tract cancer (BTC). The trial demonstrated compelling and durable clinical activity, particularly in the subgroup of patients with high HER2 expression (immunohistochemistry [IHC] 3+), with an objective response rate exceeding 50% and a median overall survival of over 18 months. These results, coupled with a manageable and tolerable safety profile devoid of chemotherapy-associated toxicities, address a profound unmet medical need in a patient population with historically poor outcomes.
Based on the strength of these data, zanidatamab secured Accelerated Approval from the U.S. Food and Drug Administration (FDA) and Conditional Marketing Authorisation from the European Medicines Agency (EMA) for the treatment of previously treated, unresectable or metastatic HER2-positive (IHC 3+) BTC. Its development is supported by a robust and ambitious clinical program aimed at expanding its use into the first-line settings for both BTC and gastroesophageal adenocarcinoma (GEA), as well as into other HER2-expressing solid tumors such as breast and colorectal cancer. This strategic pipeline positions zanidatamab not only as a new standard of care in its initial indication but also as a potential cornerstone therapy across a broad spectrum of HER2-driven malignancies. The global development and commercialization are managed through a strategic partnership between Zymeworks, the innovator, Jazz Pharmaceuticals (U.S., Europe, Japan), and BeiGene (Asia-Pacific).
Zanidatamab is an innovative antineoplastic agent that has emerged as a first-in-class targeted therapy for HER2-expressing cancers. Its development marks a significant step forward in protein engineering, designed to overcome limitations of existing HER2-targeted therapies by employing a novel bispecific binding mechanism.
Zanidatamab is the internationally recognized nonproprietary name (INN) for the molecule.[1] In the United States, it is formally identified by the nonproprietary name zanidatamab-hrii, with the four-letter, non-meaningful suffix "-hrii" used by the FDA to distinguish it from other potential future biologic products.[1] The "-hrii" suffix is a regulatory convention specific to the U.S. market, designed to enhance pharmacovigilance and ensure accurate product tracing, but it is not part of the global INN. During its development, the compound was known by the code ZW25.[1] Commercially, it is marketed under the brand name Ziihera®.[1]
From a classification standpoint, zanidatamab is a biotech therapeutic, specifically categorized as a humanized, bispecific HER2-directed monoclonal antibody.[1] Its therapeutic function places it in the pharmacologic class of antineoplastic agents. The World Health Organization has assigned it the Anatomical Therapeutic Chemical (ATC) code L01FD07.[1]
Zanidatamab is a complex protein therapeutic produced through recombinant DNA technology in Chinese Hamster Ovary (CHO) cells, a standard mammalian cell line for the production of therapeutic antibodies.[1] It is a humanized antibody, meaning the protein sequences were originally derived from a murine (mouse) source and subsequently modified to more closely resemble human antibodies, thereby reducing the potential for immunogenicity in patients.[1]
Structurally, zanidatamab is an IgG1-like bispecific antibody.[6] Its sophisticated architecture is a product of advanced protein engineering, specifically the Azymetric™ platform. It is assembled from two distinct half-antibodies, resulting in an asymmetric molecule with the structure Fab-h-CH2-CH3 x scFv-h-CH2-CH3.[7] This engineered asymmetry is fundamental to its unique biological activity, enabling it to bind two different epitopes on the HER2 receptor and, critically, to engage two separate HER2 molecules simultaneously—a capability not found in naturally occurring antibodies or conventional monoclonal antibody therapies.
The molecular formula for zanidatamab is C5553H8526N1482O1726S36, with a calculated molar mass of approximately 124,818.10 g·mol−1, or 124.8 kDa.[1] Other sources report a molecular weight of approximately 148.28 kDa, a variation that can arise from different calculation methods or post-translational modifications such as glycosylation.[6]
Table 1: Zanidatamab Drug Profile Summary
Characteristic | Value |
---|---|
International Nonproprietary Name (INN) | zanidatamab 1 |
US Adopted Name (USAN) | zanidatamab-hrii 1 |
Brand Name | Ziihera® 1 |
Development Code | ZW25 1 |
Drug Type | Biotech, Protein-Based Therapy 1 |
Drug Class | Antineoplastic, Bispecific HER2-Directed Antibody 1 |
ATC Code | L01FD07 1 |
DrugBank ID | DB15471 1 |
CAS Number | 2169946-15-8 1 |
Source | Humanized (from mouse), produced in CHO cells 1 |
Molecular Formula | C5553H8526N1482O1726S36 1 |
Molar Mass | 124,818.10 g·mol−1 1 |
The therapeutic efficacy of zanidatamab is rooted in its unique and multifaceted mechanism of action, which is a direct consequence of its engineered bispecific design. It targets the human epidermal growth factor receptor 2 (HER2, also known as ERBB2), a receptor tyrosine kinase that is a well-established oncogenic driver in a variety of solid tumors, including biliary tract, breast, and gastric cancers.[5] Aberrant HER2 signaling, typically resulting from gene amplification and subsequent protein overexpression, promotes uncontrolled cell proliferation, survival, and differentiation.[5] Zanidatamab was designed to inhibit HER2 signaling more effectively than existing agents by engaging the receptor in a novel manner.
Zanidatamab is a biparatopic antibody, meaning it is engineered to simultaneously bind two distinct and non-overlapping epitopes on the extracellular domain (ECD) of the HER2 protein.[1] This dual engagement is a key point of differentiation from monospecific antibodies. Specifically, zanidatamab targets:
By binding to both of these critical sites, zanidatamab can physically obstruct the receptor's ability to dimerize with other HER family members, a crucial step for the activation of downstream signaling pathways.[5]
The most innovative aspect of zanidatamab's mechanism is not just its dual-epitope targeting but how it engages these epitopes. Preclinical structural and biophysical studies, including surface plasmon resonance (SPR) and cryo-electron microscopy, have demonstrated that zanidatamab binds to HER2 in trans.[7] This means that a single zanidatamab molecule crosslinks two adjacent HER2 molecules on the cell surface by binding to ECD2 on one receptor and ECD4 on the other. This mode of binding is a consequence of its engineered structure, which creates a spatial constraint that prevents it from binding to two epitopes on the same HER2 molecule (
cis-binding).[9]
This trans-binding action forces a novel reorganization of HER2 receptors on the tumor cell surface. It induces the formation of large, high-order HER2 clusters and promotes the migration of these clusters into polarized "caps" on the cell surface.[7] This dramatic alteration of receptor topography is a unique feature of zanidatamab and is not observed with treatment by trastuzumab, pertuzumab, or even the combination of the two.[9]
The formation of these large HER2-zanidatamab complexes serves as a potent signal for the cell's endocytic machinery. This leads to the rapid and superior internalization of the HER2 receptor from the cell surface into the cell's interior, followed by trafficking to lysosomes for degradation.[5] The consequence is a profound depletion and downregulation of total HER2 protein levels on and within the tumor cell.[2] By physically removing the oncogenic driver from the cell surface, zanidatamab achieves a durable blockade of downstream signaling pathways, such as the MAPK pathway, thereby inhibiting tumor cell growth and promoting apoptosis.[5]
Beyond direct signal blockade, zanidatamab's IgG1 Fc domain is fully functional and engineered to recruit the patient's immune system to attack the tumor. The unique receptor clustering induced by zanidatamab creates an ideal platform for immune effector functions, leading to several forms of immune-mediated cell death:
The convergence of these multiple, synergistic mechanisms—direct signal blockade via receptor downregulation and a multi-pronged immune attack—underpins the potent anti-tumor activity of zanidatamab. This multi-modal approach may be particularly effective at overcoming the mechanisms of resistance that can limit the efficacy of monospecific antibodies like trastuzumab.[9] It is also important to note that zanidatamab is not a T-cell engager and therefore does not activate cytotoxic T-cells in a way that leads to cytokine release syndrome (CRS), a significant safety advantage over some other classes of bispecific antibodies.[13]
The pharmacokinetic profile of zanidatamab is characteristic of a large protein therapeutic, defined by intravenous administration, limited distribution, slow clearance, and a long half-life, which collectively support a convenient dosing schedule for patients.
Based on clinical trial data, the recommended dosage of zanidatamab is 20 mg/kg administered as an intravenous infusion every two weeks until disease progression or unacceptable toxicity.[15] To mitigate the risk of infusion-related reactions (IRRs), a common adverse event with monoclonal antibody therapies, premedication is recommended. Patients should receive acetaminophen, an antihistamine (e.g., diphenhydramine), and a corticosteroid (e.g., dexamethasone) approximately 30 to 60 minutes prior to each infusion.[18]
The regulatory approvals for zanidatamab were primarily based on the compelling efficacy and safety data from the HERIZON-BTC-01 study, a pivotal trial that established a new standard of care for a specific, biomarker-defined population of patients with advanced biliary tract cancer.
HERIZON-BTC-01 was a global, multicenter, open-label, single-arm, Phase 2b clinical trial designed to evaluate the efficacy and safety of zanidatamab monotherapy.[1] The study enrolled patients with HER2-amplified, unresectable, locally advanced, or metastatic biliary tract cancer (including cholangiocarcinoma and gallbladder cancer) who had experienced disease progression after receiving at least one prior gemcitabine-containing chemotherapy regimen.[1] This represents a patient population with a high unmet medical need and historically poor prognosis.
A total of 87 patients were enrolled and assigned to one of two cohorts based on centrally confirmed HER2 protein expression levels as determined by immunohistochemistry (IHC):
The primary endpoint of the study was the confirmed objective response rate (cORR) in Cohort 1, as assessed by an independent central review (ICR) according to RECIST v1.1 criteria.[15]
The HERIZON-BTC-01 trial successfully met its primary endpoint, demonstrating robust and durable anti-tumor activity. With a median follow-up of 21.9 months, the key efficacy results for the overall HER2-positive population (Cohort 1) were as follows:
A pre-specified subgroup analysis revealed that the clinical benefit of zanidatamab was most pronounced in patients with the highest level of HER2 expression (IHC 3+). This finding is of paramount clinical significance, as it validates the drug's mechanism of action, which is dependent on a high density of HER2 targets on the tumor cell surface. The efficacy in this subgroup of 62 patients was markedly superior:
These data strongly support the use of the companion diagnostic test to select patients most likely to derive substantial benefit from zanidatamab, justifying the specific labeling granted by regulatory authorities for the IHC 3+ population. When benchmarked against historical controls for second-line chemotherapy in BTC, which typically yield response rates in the single digits (around 5-8%) and a median OS of only 6 to 9 months, the results for zanidatamab in the IHC 3+ population represent a paradigm shift in treatment and a major clinical advancement.[16] This substantial improvement over available therapy provided the rationale for the FDA's Breakthrough Therapy designation and the subsequent accelerated approval.
Beyond objective tumor measurements, the clinical benefit of zanidatamab was also reflected in patient-reported outcomes from the HERIZON-BTC-01 trial. Patients who achieved a response (CR or PR) to zanidatamab generally reported clinically meaningful improvements in pain scores and less interference from pain with their daily activities compared to baseline. Furthermore, these responders had a significantly lower rate of increased opioid use (3.1%) compared to patients whose disease progressed (34.8%), indicating a tangible improvement in quality of life.[30]
Table 2: Summary of Efficacy Results from the HERIZON-BTC-01 Trial
Efficacy Endpoint | All HER2-Positive (Cohort 1, n=80) | IHC 3+ Subgroup (n=62) |
---|---|---|
Confirmed Objective Response Rate (cORR) | 41.3% 15 | 51.6% - 52% 3 |
Median Duration of Response (DOR) | 14.9 months 22 | 14.9 months 3 |
Median Overall Survival (OS) | 15.5 months 24 | 18.1 months 16 |
Median Progression-Free Survival (PFS) | 5.5 months 22 | 7.2 months 22 |
The safety profile of zanidatamab has been well-characterized through its clinical development program, particularly in the HERIZON-BTC-01 trial. The overall assessment is that the drug is generally well-tolerated, with a manageable safety profile consistent with the class of HER2-targeted therapies.[12] A key advantage is the absence of chemotherapy-associated toxicities, such as significant myelosuppression, peripheral neuropathy, or alopecia, making it an attractive option for patients who have already been exposed to cytotoxic regimens.[25]
The prescribing information for Ziihera includes several important warnings and precautions that require careful monitoring and management by healthcare professionals.
The management of zanidatamab-related adverse events is generally supportive and follows established clinical practice guidelines.
Table 3: Summary of Common and Serious Treatment-Related Adverse Events (HERIZON-BTC-01, N=87)
Adverse Event | Incidence (All Grades, %) | Incidence (Grade ≥3, %) |
---|---|---|
Diarrhea | 37% - 40% 26 | 4.6% - 5% 15 |
Infusion-Related Reaction | 33% - 35% 26 | 1.1% 25 |
Fatigue | 24% 35 | 2.3% 21 |
Abdominal Pain | 29% 35 | Not specified |
Decreased Ejection Fraction | 10.3% 17 | 3.4% 25 |
Nausea | 9.2% - 18% 25 | 1.1% 25 |
Increased AST | 6.9% - 47% 25 | 2.3% 25 |
Increased ALT | 6.9% - 46% 25 | 1.1% - 2.3% 21 |
Zanidatamab has successfully navigated the regulatory pathways in major global markets, achieving landmark approvals based on the strength of its pivotal clinical data and its potential to address a significant unmet medical need. The parallel use of expedited programs in the United States and Europe highlights a global regulatory consensus on the drug's favorable risk-benefit profile for its initial indication. This strategy allows for earlier patient access while the company completes a confirmatory Phase 3 trial to secure full approval.
The global reach of zanidatamab is expanding beyond the U.S. and Europe. A marketing application was submitted to China's National Medical Products Administration (NMPA), and conditional approval was granted in May 2025 for the treatment of previously treated HER2-positive BTC.[8] Full market availability in China, managed by BeiGene, is anticipated in the latter half of 2025.[8]
With initial approvals secured for second-line biliary tract cancer, the developers of zanidatamab have embarked on a highly ambitious and strategic clinical development program. This program is designed not only to confirm its benefit in BTC but also to expand its use into earlier lines of therapy and into a broad range of other HER2-expressing solid tumors. The overarching goal is to establish zanidatamab as a foundational therapy for HER2-driven cancers, potentially displacing existing standards of care.
This ongoing, global, randomized, open-label Phase 3 trial is the mandatory confirmatory study required by both the FDA and EMA. It is evaluating the efficacy and safety of zanidatamab in combination with standard-of-care (SoC) therapy (cisplatin and gemcitabine, with or without a PD-1/L1 inhibitor) versus SoC therapy alone in the first-line setting for patients with advanced HER2-positive BTC.[8] A positive outcome in this trial would secure full regulatory approval and potentially move zanidatamab into the first-line treatment paradigm.
This is a large-scale, global, randomized, open-label Phase 3 trial expected to enroll approximately 900 patients. It is designed to directly challenge the current standard of care, trastuzumab. The study is evaluating zanidatamab plus chemotherapy against trastuzumab plus chemotherapy as a first-line treatment for patients with HER2-positive advanced or metastatic gastroesophageal adenocarcinoma (GEA), which includes stomach, gastroesophageal junction, and esophageal cancers.[40] A third experimental arm is investigating the triplet combination of zanidatamab, chemotherapy, and tislelizumab (an anti-PD-1 antibody), exploring the potential synergy of dual HER2 blockade with immune checkpoint inhibition.[41] This trial leverages the corporate partnership with BeiGene, the developer of tislelizumab, and if successful, could establish a new, potentially superior standard of care in a major oncology indication.
Zanidatamab is being actively investigated in HER2-positive breast cancer, a setting with multiple established therapies but still significant unmet needs.
Reflecting a modern, biomarker-driven approach to oncology, the DiscovHER PAN-206 trial is a Phase 2 "basket" study. It is designed to evaluate the efficacy of zanidatamab monotherapy across a wide range of locally advanced or metastatic solid tumors that are HER2-positive (defined as IHC 3+), with the exception of BTC. This includes, but is not limited to, colorectal, endometrial, non-small cell lung, ovarian, urothelial, and pancreatic cancers.[39] Success in this trial could support a tumor-agnostic approval, significantly broadening zanidatamab's potential patient population based solely on the presence of the HER2 IHC 3+ biomarker.
Table 4: Overview of Major Ongoing Clinical Trials for Zanidatamab
Trial Name (NCT ID) | Phase | Indication | Setting | Comparator(s) | Strategic Goal |
---|---|---|---|---|---|
HERIZON-BTC-302 (NCT06282575) | 3 | Biliary Tract Cancer | 1st Line | Standard of Care (Chemo +/- PD-1/L1) 40 | Convert accelerated/conditional approval to full approval; establish 1L standard of care. |
HERIZON-GEA-01 (NCT05152147) | 3 | Gastroesophageal Adenocarcinoma | 1st Line | Trastuzumab + Chemo 41 | Displace trastuzumab as 1L standard of care; explore synergy with PD-1 inhibition. |
EMPOWHER-303 (NCT06435429) | 3 | Breast Cancer | Metastatic (Post-T-DXd) | Trastuzumab + Chemo 40 | Address unmet need in patients progressing on modern ADCs. |
DiscovHER PAN-206 (NCT06695845) | 2 | Pan-Tumor (Solid Tumors) | Previously Treated | Single-Arm | Establish broad, tumor-agnostic activity in HER2 IHC 3+ cancers for label expansion. |
Zanidatamab has firmly established itself as a practice-changing therapy and the undisputed standard of care for adult patients with previously treated, unresectable or metastatic HER2-positive (IHC 3+) biliary tract cancer. Its unique biparatopic mechanism of action translates into superior preclinical activity and clinically meaningful outcomes in a patient population with a dire prognosis and limited therapeutic options. The combination of a high objective response rate, durable responses, a significant overall survival benefit, and a manageable, chemotherapy-free safety profile provides a compelling clinical rationale for its use. The convenient bi-weekly dosing schedule further enhances its attractiveness for both clinicians and patients.
The future role and ultimate commercial success of zanidatamab will be defined by the outcomes of its ambitious Phase 3 clinical development program. Success in the HERIZON-GEA-01 or EMPOWHER-303 trials would be transformative, elevating zanidatamab from a successful drug for a rare cancer into a major oncology product capable of challenging long-standing standards of care in much larger indications. The pan-tumor strategy of the DiscovHER PAN-206 trial also holds the potential to unlock significant value by establishing a biomarker-driven, tumor-agnostic label.
The global strategy for zanidatamab is managed through a tripartite corporate structure that leverages the strengths of each partner. Zymeworks serves as the innovator and licensor of the core technology. Jazz Pharmaceuticals has secured the development and commercialization rights for the major markets of the United States, Europe, and Japan, and has successfully guided the drug through its initial regulatory approvals.[8] BeiGene holds the exclusive rights for development and commercialization in key Asia-Pacific territories, including China, Australia, and New Zealand, and is also a strategic partner in the combination study with its PD-1 inhibitor, tislelizumab.[8] This collaborative framework ensures a coordinated global development strategy and broad market access.
Despite the clear success of zanidatamab, several important questions remain that will be addressed through ongoing research and long-term follow-up. A primary focus will be understanding the mechanisms of acquired resistance that will inevitably emerge. Identifying biomarkers of response and resistance beyond HER2 IHC status will be crucial for optimizing patient selection and developing rational next-line combination strategies. The long-term durability of responses and the ultimate impact on survival will continue to be monitored in all ongoing trials. Finally, the exploration of zanidatamab in combination with other novel agents, such as antibody-drug conjugates (ADCs), other targeted therapies, or next-generation immunotherapies, will be essential to further improve outcomes and fully define zanidatamab's legacy in the comprehensive treatment of HER2-positive cancers.
Published at: September 12, 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.