SHR-A1904, also known by the non-proprietary name garetatug rezetecan, is an investigational antibody-drug conjugate (ADC) at the forefront of a new wave of targeted cancer therapies.[1] Developed by the Chinese pharmaceutical leader Jiangsu Hengrui Pharmaceuticals Co., Ltd., SHR-A1904 is engineered to selectively target Claudin-18 isoform 2 (CLDN18.2), a protein antigen with highly restricted expression in normal tissues that becomes exposed on the surface of various cancer cells, particularly those of gastric and gastroesophageal junction (G/GEJ) origin.[2] The agent represents a sophisticated application of ADC technology, combining a high-specificity monoclonal antibody with a potent DNA topoisomerase I inhibitor payload via a stable, cleavable linker system.[2]
The clinical development program for SHR-A1904 has been aggressive and strategically multifaceted. Foundational Phase 1 data, notably published in the peer-reviewed journal Nature Medicine, have demonstrated a manageable safety profile and encouraging anti-tumor activity in heavily pre-treated patients with CLDN18.2-positive advanced G/GEJ cancer.[2] These results have propelled the asset into late-stage, pivotal Phase 3 trials in China for stomach adenocarcinoma and related indications, positioning it for potential near-term approval in its domestic market.[2] Concurrently, Hengrui has initiated a global development strategy, with trials underway in the United States and Australia to evaluate SHR-A1904 in a broader range of advanced solid tumors.[2]
The strategic importance and perceived potential of SHR-A1904 were significantly validated in October 2023 through a major collaboration with Merck KGaA, Darmstadt, Germany. The agreement grants the global pharmaceutical giant an exclusive option to license the ex-China rights to develop, manufacture, and commercialize the ADC, in a deal potentially valued at up to €1.4 billion.[3] This partnership not only provides a substantial financial endorsement but also secures a powerful and experienced partner to navigate the complexities of global regulatory approval and commercialization.
SHR-A1904 is emerging as a leading contender in an increasingly competitive therapeutic landscape focused on CLDN18.2. While the approval of the monoclonal antibody zolbetuximab has validated the target, its modest efficacy has created a clear opportunity for more potent next-generation agents like ADCs. SHR-A1904's future trajectory will be defined by its ability to navigate several critical challenges: confirming the promising efficacy signals from its early trials in larger, randomized studies; carefully managing a safety profile characterized by hematological and gastrointestinal toxicities, particularly in combination regimens; and ultimately demonstrating a superior or differentiated clinical profile versus a growing field of direct competitors. The execution of its dual-pronged China-focused and global partnership strategy will be paramount to realizing its full therapeutic and commercial potential.
The development and strategic positioning of SHR-A1904 offer a compelling view into several key trends shaping the modern oncology landscape. The program is not merely the advancement of a single drug candidate; it is emblematic of the maturation of China's biopharmaceutical sector, the strategic evolution of ADC technology, and the sophisticated clinical development pathways required to succeed in a competitive market.
A primary theme surrounding SHR-A1904 is its role as a testament to the rise of genuine innovation within Chinese biopharma. For years, the sector was largely characterized by the development of biosimilars or "fast-follower" assets. However, Jiangsu Hengrui's journey with ADCs illustrates a significant strategic evolution. The company's engagement in this complex therapeutic modality dates back over a decade, representing a sustained and deep-seated commitment to building internal expertise.[3] The establishment of its proprietary HRMAP ADC strategic platform signifies a transition from imitation to the creation of a robust, innovation-driven R&D engine.[3] The culmination of this long-term investment is SHR-A1904, an asset developed in-house with its own intellectual property.[7] The subsequent partnership with Merck KGaA is perhaps the most critical external validation of this strategy. The agreement, with a potential value of €1.4 billion, was Hengrui's first major overseas licensing deal for an ADC and was explicitly cited as an "initial recognition of its platform".[3] This is not simply a transaction for a promising molecule but an endorsement by a major Western pharmaceutical company of the underlying technology and R&D capabilities originating from China. This mirrors a broader industry shift, where Chinese-developed ADCs accounted for over 40% of global out-licensing deals in this class in 2023, signaling a fundamental rebalancing of the global sources of pharmaceutical innovation.[5]
The molecular design of SHR-A1904, specifically its choice of a topoisomerase I (Topo-I) inhibitor payload, represents a key technical and strategic differentiator. The ADC field has been significantly influenced by the remarkable success of agents like Enhertu (trastuzumab deruxtecan) and Trodelvy (sacituzumab govitecan), both of which utilize Topo-I inhibitor payloads.[8] Hengrui’s selection of a novel, lipophilicity-enhanced Topo-I inhibitor, SHR9265, for SHR-A1904 aligns the asset with this proven, highly potent class of cytotoxins.[3] The enhanced lipophilicity is a deliberate design choice intended to improve the payload's ability to permeate cell membranes.[5] This property is crucial for mediating a strong "bystander effect," whereby the released payload can diffuse from the target cancer cell to kill adjacent tumor cells, even those that may have low or no CLDN18.2 expression. This mechanism has the potential to generate deeper and more durable responses in heterogenous tumors. This strategic choice to maximize cytotoxic potency differentiates SHR-A1904 from competitors utilizing different payload classes, such as auristatins (e.g., AZD0901), which have distinct mechanisms of action and toxicity profiles.[9] This decision, however, also carries implications for the drug's safety profile, as Topo-I inhibitor payloads are often associated with hematological toxicities, creating a critical therapeutic index that must be carefully managed throughout clinical development.
Finally, Hengrui is pursuing a remarkably aggressive and multi-pronged clinical development strategy that reflects high confidence in the asset and a sophisticated understanding of the global oncology market. Rather than a sequential, geographically limited approach, the company is simultaneously driving SHR-A1904 toward approval in its domestic market while laying the groundwork for global expansion and broader utility. The program features parallel tracks: late-stage Phase 3 monotherapy trials aimed at securing approval for second-line G/GEJ cancer in China [2], and concurrent Phase 2 studies in the United States and Australia targeting a wider indication of "Advanced Malignant Solid Neoplasm" to generate data for Western regulatory bodies.[2] Most ambitiously, Hengrui has initiated a large and complex Phase 1b/3 platform trial (NCT06350006) to proactively evaluate SHR-A1904 in combination with both standard-of-care chemotherapy (CAPOX) and immunotherapy (the anti-PD-L1 antibody Adebrelimab).[12] This forward-looking approach does not wait for monotherapy approval before exploring combinations. It is designed to rapidly establish SHR-A1904's potential as a backbone therapy in earlier lines of treatment, a strategy essential for maximizing the commercial potential of a new oncology agent. This comprehensive and well-funded strategy underscores Hengrui's ambition to position SHR-A1904 not just as another treatment option, but as a foundational therapy for CLDN18.2-positive cancers globally.
The therapeutic rationale for SHR-A1904 is anchored in the unique biology of its target, Claudin-18 isoform 2 (CLDN18.2). CLDN18.2 is a 261-amino acid transmembrane protein that serves as a critical component of tight junctions, the paracellular barriers that regulate the passage of molecules between adjacent epithelial cells and maintain cellular polarity.[14] In healthy human tissues, the expression of CLDN18.2 is highly restricted, found almost exclusively within the differentiated epithelial cells of the gastric mucosa.[2] Critically, in this non-malignant state, the extracellular domains of the CLDN18.2 protein are sequestered within the tight junction complex, making them inaccessible to circulating antibodies in the bloodstream.[2]
This biological sequestration is profoundly disrupted during the process of malignant transformation. As gastric epithelial cells become cancerous, they often lose their tight cell-to-cell architecture and polarity. This structural disruption leads to the exposure of the CLDN18.2 protein's extracellular loops on the tumor cell surface, effectively unmasking it as a cancer-specific antigen.[2] This change in accessibility, coupled with the fact that CLDN18.2 expression is often maintained or even upregulated in a significant proportion of gastric, gastroesophageal junction, and pancreatic cancers, transforms it into an ideal target for antibody-based therapies.[2] The tumor-specific presentation of the target antigen creates a large potential therapeutic window, allowing for potent on-target activity against cancer cells while minimizing the risk of on-target, off-tumor toxicity in healthy gastric tissues. This unique characteristic underpins the development of a diverse pipeline of agents targeting CLDN18.2, including monoclonal antibodies, bispecific antibodies, CAR-T cells, and ADCs like SHR-A1904.[14]
SHR-A1904 is a sophisticated, tripartite molecular construct, with each component engineered to perform a specific function in the targeted delivery of a cytotoxic agent to cancer cells.[2] The synergy between the antibody, linker, and payload is central to its mechanism of action and overall therapeutic profile.
The Antibody: The backbone of the ADC is a monoclonal antibody specifically designed to recognize and bind with high affinity to the exposed extracellular domains of CLDN18.2 on the surface of tumor cells.[2] This binding event is the critical first step, ensuring that the ADC selectively accumulates at the tumor site. Once bound to CLDN18.2, the entire antibody-drug conjugate-antigen complex is internalized by the cancer cell through the process of endocytosis, transporting the cytotoxic payload from the extracellular environment into the cell's interior.[7]
The Linker: The antibody is connected to its cytotoxic payload via a deliberately designed linker molecule. SHR-A1904 utilizes a cleavable, peptide-based linker composed of the tetrapeptide sequence Gly-Gly-Phe-Gly (GGFG).[3] This linker chemistry is crucial for the ADC's stability and safety. It is engineered to remain stable in the systemic circulation, preventing the premature release of the toxic payload, which could otherwise cause widespread systemic toxicity.[5] Only after the ADC has been internalized into the lysosomal compartment of the cancer cell is the linker exposed to specific intracellular enzymes, such as cathepsins, which are highly active in the acidic lysosomal environment. These enzymes recognize and cleave the GGFG sequence, precisely releasing the active payload at its intended site of action.[3]
The Payload (SHR9265): The cytotoxic warhead of SHR-A1904 is a novel and highly potent small molecule drug named SHR9265, which functions as a DNA topoisomerase I inhibitor.[2] Topoisomerase I is an essential nuclear enzyme that relieves torsional strain in DNA during replication and transcription by creating and then re-ligating single-strand breaks. Once released from the linker, SHR9265 intercalates into the DNA-topoisomerase I complex, stabilizing it and preventing the re-ligation step. This leads to the accumulation of irreversible DNA single-strand breaks, which, upon encountering a replication fork, are converted into catastrophic double-strand breaks. This extensive DNA damage overwhelms the cell's repair mechanisms, ultimately triggering programmed cell death, or apoptosis.[16] The molecular design of SHR9265 is described as "lipophilicity-enhanced," a feature that increases its ability to diffuse across cell membranes.[5] This property is vital for the "bystander effect," allowing the released payload to exit the targeted CLDN18.2-positive cancer cell and kill neighboring tumor cells that may have lower or no CLDN18.2 expression, thereby amplifying the ADC's anti-tumor activity within the tumor microenvironment.[5]
Component | Description | Source(s) |
---|---|---|
Drug Name(s) | SHR-A1904; garetatug rezetecan | 1 |
Drug Class | Antibody-Drug Conjugate (ADC) | 2 |
Target Antigen | Claudin-18 isoform 2 (CLDN18.2) | 2 |
Antibody Component | Anti-CLDN18.2 monoclonal antibody | 2 |
Linker Type | Cleavable, peptide-based (GGFG) | 3 |
Payload Class | DNA Topoisomerase I Inhibitor | 2 |
Payload Name | SHR9265 | 3 |
Originator | Jiangsu Hengrui Pharmaceuticals Co., Ltd. | 2 |
Global Partner (ex-China) | Merck KGaA, Darmstadt, Germany / EMD Serono (Option) | 1 |
The clinical development program for SHR-A1904 is characterized by its ambition, strategic parallelism, and global reach. Jiangsu Hengrui Pharmaceuticals has constructed a comprehensive pipeline of studies designed to rapidly establish proof-of-concept, secure regulatory approval in its domestic market, and simultaneously generate the data necessary for global expansion and combination therapy development. This multi-pronged approach reflects a high degree of confidence in the asset and a sophisticated strategy aimed at maximizing its clinical and commercial potential across multiple geographies and treatment settings.
The foundational trial for SHR-A1904 is NCT04877717, a first-in-human, open-label, three-stage Phase 1 study conducted exclusively in China.[2] This study was meticulously designed to systematically evaluate the drug's core properties. The first stage was a dose-escalation phase to determine the safety, tolerability, and dose-limiting toxicities (DLTs) of SHR-A1904 across a range of doses.[17] The subsequent stages involved pharmacokinetic (PK) and efficacy expansion cohorts, which were designed to further characterize the drug's behavior in the body and to obtain a robust preliminary signal of its anti-tumor activity in patients with CLDN18.2-positive advanced or metastatic gastric or gastroesophageal junction (G/GEJ) cancer.[2] The successful completion of this study and the subsequent publication of its results in the high-impact journal
Nature Medicine provided the critical validation and scientific credibility necessary to advance the program into later-stage development.[2]
Building on the initial success in China, Hengrui initiated NCT05277168, an open-label, single-arm, multi-center Phase 1/2a clinical study designed to support the global development of SHR-A1904.[11] This trial expands the investigation beyond G/GEJ cancer to include a broader population of patients with advanced solid tumors, explicitly including pancreatic cancer, that express CLDN18.2.[11] Crucially, this study established a clinical footprint outside of China, with trial sites activated in the United States and Australia.[2] The primary objectives of this study are to assess the safety and tolerability of SHR-A1904 in a more diverse patient population and to determine the maximum tolerated dose (MTD) and/or the recommended Phase 2 dose (RP2D) for future global studies.[11] This trial is a linchpin in the ex-China strategy, as the data generated will be essential for regulatory submissions to agencies such as the U.S. Food and Drug Administration (FDA).
Perhaps the most ambitious component of the development program is NCT06350006, a large-scale (planned enrollment of 924 patients), randomized, multi-center study investigating SHR-A1904 in combination regimens for CLDN18.2-positive advanced solid tumors.[12] This trial's sophisticated design reflects a forward-looking strategy to position SHR-A1904 as a cornerstone of treatment. The study is divided into two main parts:
In parallel with its global and combination therapy efforts, Hengrui is aggressively advancing SHR-A1904 as a monotherapy in its domestic market. The drug has progressed to Phase 3 clinical trials in China for several key indications, including second-line or greater treatment for stomach adenocarcinoma, CLDN18.2-positive G/GEJ adenocarcinoma, and CLDN18.2-positive stomach cancer.[2] This focused approach leverages the strong initial data from the Phase 1 trial and represents the most direct and rapid path to commercialization for the drug, aiming to address the high unmet need for effective therapies for these cancers within the Chinese population.
Trial ID (NCT) | Phase | Title / Brief Description | Patient Population | Intervention(s) | Status | Geography |
---|---|---|---|---|---|---|
NCT04877717 | Phase 1 | A Study in Advanced G/GEJ Cancer | CLDN18.2+ G/GEJ Cancer | SHR-A1904 Monotherapy | Active, not recruiting | China |
NCT05277168 | Phase 1/2a | A Study in Advanced Solid Tumors | CLDN18.2+ Solid Tumors (incl. Pancreatic) | SHR-A1904 Monotherapy | Recruiting | United States, Australia |
NCT06350006 | Phase 1b/3 | SHR-A1904 Combinations Study | CLDN18.2+ Solid Tumors | SHR-A1904 + Adebrelimab +/- CAPOX | Recruiting | China |
N/A | Phase 3 | Monotherapy in 2L+ G/GEJ Cancer | CLDN18.2+ G/GEJ Cancer | SHR-A1904 Monotherapy | Active | China |
The first-in-human Phase 1 trial, NCT04877717, serves as the clinical foundation for the entire SHR-A1904 development program. The results, presented at major oncology conferences and published in Nature Medicine, have provided a detailed initial assessment of the drug's efficacy and safety, guiding dose selection and future trial design.[2]
NCT04877717 was an open-label, three-stage study conducted at multiple centers across China.[2] The trial enrolled a total of 95 patients with pathologically confirmed, CLDN18.2-positive, locally advanced unresectable or metastatic gastric or gastroesophageal junction cancer.[2] A key characteristic of the study population was that they were heavily pre-treated, having progressed on or been intolerant to available standard therapies, representing a patient group with a high unmet medical need and historically poor prognosis.[2] The study's three-part design included a dose-escalation phase (with doses ranging from 0.6 mg/kg to 8.0 mg/kg), followed by pharmacokinetic and efficacy expansion phases at selected dose levels.[2]
The efficacy results from NCT04877717 have been reported across multiple time points and patient subsets, painting a picture of promising and evolving anti-tumor activity. A comprehensive analysis requires synthesizing these different data cuts.
The primary publication in Nature Medicine, with a data cutoff of November 1, 2024, provided the most extensive initial dataset on 95 patients. In this analysis, the 6.0 mg/kg and 8.0 mg/kg dose levels were selected for the expansion cohorts.[4] At the 6.0 mg/kg dose, the confirmed objective response rate (cORR) was 24.2% (95% CI, 11.1–42.3), and the median progression-free survival (PFS) was 5.6 months (95% CI, 3.0–6.9). At the 8.0 mg/kg dose, the results were similar, with a cORR of 25.0% (95% CI, 12.1–42.2) and a median PFS of 5.8 months (95% CI, 3.0–8.6).[4]
Data presented by Jiangsu Hengrui, summarized by Larvol, provided further details on 74 evaluable patients. This report showed an overall ORR of 35.1% and a cORR of 23.0%.[1] The dose-specific cORRs were 26.7% for 6.0 mg/kg and 26.5% for 8.0 mg/kg, closely aligning with the
Nature Medicine findings. A particularly encouraging metric from this dataset was the median duration of response (DoR), which was not reached in the 6.0 mg/kg group (95% CI, 4.2 months–not reached) and was a robust 8.1 months in the 8.0 mg/kg group (95% CI, 3.9–not reached), suggesting that the responses, when they occurred, were durable.[1]
A later data cut, with a cutoff date of March 18, 2024, was discussed by lead investigator Dr. Ruihua Xu and reported by OncLive. This analysis, focused on smaller subsets of efficacy-evaluable patients, showed a notable improvement in response rates, especially at the lower of the two expansion doses.[18] Among nine patients in the 6.0 mg/kg cohort, the ORR reached 55.6% (95% CI, 21.2%-86.3%), with a disease control rate (DCR) of 88.9% (95% CI, 51.8%-99.7%). In the larger 8.0 mg/kg cohort of 30 patients, the ORR was 36.7% (95% CI, 19.9%-56.1%), with a DCR of 86.7% (95% CI, 69.3%-96.2%).[18]
The divergence between the earlier, larger dataset (cORR ~25%) and the later, smaller cohort data (ORR 55.6% at 6.0 mg/kg) is a critical analytical point. While the higher ORR in the later update could be influenced by statistical variability in a small subgroup (n=9), it may also reflect a maturing of responses with longer follow-up or a refinement in patient selection within the expansion cohort. The superior performance observed at the lower 6.0 mg/kg dose is particularly significant. It suggests that this dose may occupy a "sweet spot" on the therapeutic index curve, delivering potent efficacy with a more favorable safety profile compared to the higher dose. This observation is crucial for the drug's future, as a better-tolerated yet highly effective dose is more suitable for combination strategies and has a higher probability of regulatory and clinical success. The clinical importance of these findings was underscored by Dr. Xu's commentary that these response rates "may be the highest that have been achieved so far in this difficult-to-treat patient population".[18]
The safety profile of SHR-A1904 is consistent with its mechanism as an ADC carrying a potent Topo-I inhibitor payload, characterized primarily by hematological and gastrointestinal adverse events. The Phase 1 trial provided a detailed look at its tolerability and established a dose-dependent toxicity pattern.
Dose-limiting toxicities (DLTs) were observed during the dose-escalation phase. At the 4.8 mg/kg dose level, two patients experienced DLTs: one with Grade 3 febrile neutropenia and another with Grade 3 increased blood bilirubin. At the 6.0 mg/kg level, one DLT of Grade 3 gastric mucosal lesion was reported.[4] Despite these events, the maximum tolerated dose (MTD) was not formally reached within the tested range, indicating that the observed toxicities were considered manageable by the investigators, leading to the selection of the 6.0 mg/kg and 8.0 mg/kg doses for further study.[4]
Across all 95 patients in the Nature Medicine report, treatment-emergent adverse events (TEAEs) of any grade occurred in 100% of participants.[4] The most frequently reported TEAEs were anemia (75.8%), nausea (67.4%), hypoalbuminemia (64.2%), and decreased white blood cell count (58.9%). A majority of patients (62.1%) experienced drug-related Grade 3 or higher adverse events, and importantly, no treatment-related deaths were reported.[4]
The dose-dependency of the toxicity became clearer in the analysis of the expansion cohorts. Data reported by OncLive showed a significant difference in the rate of severe adverse events between the two dose levels. Among safety-evaluable patients, Grade 3 or higher treatment-related adverse events (TRAEs) occurred in just 26.7% of those receiving the 6.0 mg/kg dose. This rate increased dramatically to 71.1% in patients receiving the 8.0 mg/kg dose.[18] This stark contrast in severe toxicity strongly reinforces the clinical rationale for focusing on the 6.0 mg/kg dose. When combined with the efficacy data suggesting potentially superior activity at this lower dose, a compelling case emerges for 6.0 mg/kg offering an optimized therapeutic index. This favorable balance of efficacy and safety is a critical attribute for any new oncology drug, particularly for one intended for use in combination regimens where overlapping toxicities with other agents are a primary concern. The overall safety profile was deemed "manageable," a positive conclusion for an ADC of this class.[1]
Metric | 6.0 mg/kg Dose | 8.0 mg/kg Dose | Source(s) |
---|---|---|---|
cORR (Nature Medicine) | 24.2% (95% CI, 11.1–42.3) | 25.0% (95% CI, 12.1–42.2) | 4 |
cORR (Press Release) | 26.7% (95% CI, 12.3–45.9) | 26.5% (95% CI, 12.9–44.4) | 1 |
ORR (OncLive) | 55.6% (95% CI, 21.2%-86.3%) | 36.7% (95% CI, 19.9%-56.1%) | 18 |
DCR (OncLive) | 88.9% (95% CI, 51.8%-99.7%) | 86.7% (95% CI, 69.3%-96.2%) | 18 |
Median PFS (Nature Medicine) | 5.6 months (95% CI, 3.0–6.9) | 5.8 months (95% CI, 3.0–8.6) | 4 |
Median DoR (Press Release) | Not Reached (95% CI, 4.2–NR) | 8.1 months (95% CI, 3.9–NR) | 1 |
Adverse Event | Any Grade (%) | Grade ≥3 (%) | Source(s) |
---|---|---|---|
Anemia | 75.8% | N/A | 4 |
Nausea | 67.4% | N/A | 4 |
Hypoalbuminemia | 64.2% | N/A | 4 |
Decreased White Blood Cell Count | 58.9% | N/A | 4 |
All Drug-Related AEs | 100% | 62.1% | 4 |
Grade ≥3 TRAEs at 6.0 mg/kg | 86.7% | 26.7% | 18 |
Grade ≥3 TRAEs at 8.0 mg/kg | 94.7% | 71.1% | 18 |
SHR-A1904 is not an isolated project but rather the flagship asset emerging from a deep and long-term strategic commitment by Jiangsu Hengrui Pharmaceuticals to become a leader in the field of antibody-drug conjugates.[3] The company's involvement in ADC development is not a recent pivot but a sustained effort that began as early as 2011. This decade-plus journey has seen Hengrui evolve from initial explorations of analogues to established first-generation ADCs to the creation of its own proprietary third-generation ADC technology platform, known as HRMAP.[3] This platform underpins a broad and diversified pipeline that includes at least 12 distinct ADC candidates in various stages of clinical development.[3]
Beyond SHR-A1904, Hengrui's ADC portfolio targets a range of validated and novel cancer antigens, including SHR-A2009 (targeting HER3) and SHR-A2102 (targeting Nectin-4).[3] This extensive pipeline demonstrates a corporate strategy that views ADCs as a core pillar of its future oncology franchise. By investing heavily in platform technology and advancing multiple candidates in parallel, Hengrui is mitigating the risk associated with any single asset and building a formidable presence in one of the most dynamic and promising areas of cancer therapy. The development of SHR-A1904, therefore, should be seen in this broader context: it is the most advanced product of a mature, well-funded, and strategically focused R&D engine, intended to be the first of many innovative ADCs that the company brings to the global market.
A pivotal moment in the development and strategic validation of SHR-A1904 occurred in October 2023, when Jiangsu Hengrui announced a major strategic collaboration with the German pharmaceutical company Merck KGaA, Darmstadt, Germany.[3] The agreement was structured around two of Hengrui's innovative oncology assets. The primary component was an exclusive license for Merck KGaA to develop, manufacture, and commercialize Hengrui's novel PARP1 inhibitor, HRS-1167, in all territories outside of mainland China. Critically, the deal also included an exclusive
option for Merck KGaA to license SHR-A1904 under the same global ex-China terms.[5]
The financial terms of the agreement are substantial, reflecting the high value placed on Hengrui's technology. Merck KGaA made an upfront payment of €160 million, with potential future payments for development, regulatory, and commercial milestones, plus tiered royalties on net sales, that could bring the total deal value to €1.4 billion.[3]
This partnership is a strategically astute move for both companies, creating a symbiotic relationship that de-risks the global development of SHR-A1904. For Jiangsu Hengrui, the deal provides a significant infusion of non-dilutive capital and, more importantly, secures a top-tier global partner with the extensive experience, infrastructure, and resources required to navigate the complex and costly pathways of global clinical trials, regulatory submissions, and commercial launches. This allows Hengrui to focus its own resources on dominating the large and rapidly growing Chinese market, where it has a home-field advantage.
For Merck KGaA, the deal structure represents a shrewd, low-risk entry into the highly competitive CLDN18.2 ADC space. By acquiring an option rather than an outright license, Merck KGaA has the flexibility to wait for more mature clinical data from both SHR-A1904 and its competitors before committing the full financial and operational resources needed for late-stage development. At the same time, the option effectively takes a highly promising asset off the market, preventing a direct competitor from acquiring it. The strategic fit is also strong. As articulated by Merck KGaA's Global Head of Research & Development, Danny Bar-Zohar, and supported by industry analysis, SHR-A1904 diversifies the company's oncology pipeline in its focus areas of DNA damage response and ADCs.[5] It complements Merck KGaA's internal ADC portfolio, which utilizes different linker-payload technologies, such as their CEACAM5-targeting ADC, M9140.[5] Given this strong strategic alignment and the promising early data for SHR-A1904, there is a high probability that Merck KGaA will exercise its option, setting the stage for a major global push for the asset.
SHR-A1904 is entering one of the most dynamic and crowded development areas in oncology. The validation of CLDN18.2 as a therapeutic target has catalyzed an "overflowing" pipeline of investigational agents, including monoclonal antibodies, ADCs, bispecific antibodies, and CAR-T cell therapies, all vying for a share of the significant market opportunity in gastric, pancreatic, and other solid tumors.[14] To succeed, SHR-A1904 must not only demonstrate safety and efficacy but also establish a clear point of differentiation within this competitive field.
The first agent to successfully target CLDN18.2 and gain regulatory approval is zolbetuximab (brand name Vyloy), developed by Astellas.[9] It is crucial to note that zolbetuximab is a chimeric monoclonal antibody, not an ADC.[27] Its mechanism of action does not involve the delivery of a cytotoxic payload but rather relies on the immune system to kill tumor cells through processes like antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC).[27]
Zolbetuximab received its first approval in Japan, followed by U.S. FDA approval in October 2024, for the first-line treatment of patients with CLDN18.2-positive, HER2-negative, locally advanced unresectable or metastatic G/GEJ adenocarcinoma, in combination with standard chemotherapy regimens like mFOLFOX6 or CAPOX.[9] Approval was based on the results of two pivotal Phase 3 trials, SPOTLIGHT and GLOW.[31] These studies demonstrated a statistically significant improvement in both progression-free survival (PFS) and overall survival (OS) for the zolbetuximab-plus-chemotherapy arms compared to chemotherapy alone. However, the magnitude of the benefit, while clinically meaningful, was modest. For example, in the SPOTLIGHT trial, the addition of zolbetuximab to mFOLFOX6 improved median PFS from 8.7 months to 10.6 months and median OS from 15.5 months to 18.2 months.[32]
The approval of zolbetuximab is a landmark event that has established CLDN18.2 as a clinically validated and commercially viable target. However, its modest efficacy gains set a clear bar for next-generation therapies. This creates a significant opportunity for more potent modalities like ADCs. Any ADC, including SHR-A1904, will be expected to deliver substantially better efficacy outcomes—such as higher response rates and longer survival—to justify its adoption in clinical practice, particularly given that ADCs often come with more complex safety and tolerability profiles than naked monoclonal antibodies.
SHR-A1904 faces its most direct competition from a cohort of other CLDN18.2-targeted ADCs that are also advancing rapidly through clinical development. These agents differ primarily in their choice of cytotoxic payload and the specifics of their linker and antibody design, which may translate into different clinical profiles.
Other ADCs in earlier stages of development include SYSA1801, LM-302, and SOT102, ensuring that the competitive landscape will remain crowded and dynamic for the foreseeable future.[9]
Drug Name | Developer(s) | Payload Type | Highest Development Phase | Key Efficacy Data (G/GEJ Cancer) | Key Differentiator / Strategic Note |
---|---|---|---|---|---|
SHR-A1904 | Jiangsu Hengrui / Merck KGaA (Option) | Topoisomerase I Inhibitor | Phase 3 | cORR ~25%; ORR up to 55.6% in subgroup | Strong early efficacy signal, especially at 6.0 mg/kg dose; major global partner. |
IBI-343 | Innovent Biologics | Topoisomerase I Inhibitor | Phase 3 | cORR 47.1% (high expressors) | Very direct competitor with similar mechanism and strong efficacy data. |
AZD0901 | AstraZeneca / Keymed Biosciences | Auristatin (MMAE) | Phase 3 | Promising Phase 1 activity | Different payload class (microtubule inhibitor); backed by AstraZeneca. |
RC118 | RemeGen | Auristatin (MMAE) | Phase 1/2 | Promising Phase 1/2 activity | Different payload class; FDA Orphan Drug Designations for GC and PC. |
MK-1200 | Merck & Co / Kelun-Biotech | Topoisomerase I Inhibitor | Phase 1/2 | Data not yet mature | Similar mechanism to SHR-A1904; backed by global leader Merck & Co. |
SHR-A1904 (garetatug rezetecan) has emerged as a highly promising and strategically significant asset in the oncology pipeline. Its development by Jiangsu Hengrui Pharmaceuticals represents a landmark achievement for the Chinese biopharmaceutical industry, showcasing a transition from fast-following to genuine, globally competitive innovation. The drug's sophisticated design, which pairs a validated, tumor-specific target with a potent Topo-I inhibitor payload, has translated into compelling early clinical results, demonstrating significant anti-tumor activity in a difficult-to-treat patient population. The substantial partnership with Merck KGaA provides a clear and powerful pathway for global development, positioning SHR-A1904 as a leading contender in the race to define the next generation of therapy for CLDN18.2-positive cancers.
The trajectory of SHR-A1904 from a promising clinical candidate to a standard-of-care therapy will depend on its ability to successfully navigate several key milestones and answer critical questions over the next several years.
First and foremost is the confirmation of its efficacy in ongoing and future Phase 3 trials. The central question is whether the remarkable 55.6% ORR observed at the 6.0 mg/kg dose can be replicated in a larger, randomized setting. The outcome of the head-to-head comparison against standard chemotherapy in the second-line setting in China will be the first major test.
Second, the performance of SHR-A1904 in combination regimens will be a crucial determinant of its ultimate market size. The large NCT06350006 trial will be closely watched to see if the addition of SHR-A1904 to chemotherapy and immunotherapy yields a synergistic effect on efficacy and whether the combined toxicity profile remains manageable. Success in this trial could position SHR-A1904 as a first-line therapy.
Third, the competitive dynamics will continue to evolve rapidly. As data from competing ADCs like IBI-343 and AZD0901 mature, a clearer picture of the relative strengths and weaknesses of different payload technologies and molecular designs will emerge. It remains to be seen whether one agent will establish clear superiority or if different ADCs will find distinct clinical niches based on factors like patient biomarker status, line of therapy, or tolerability.
Finally, the strategic partnership with Merck KGaA will move to the forefront. The timing and decision of Merck KGaA to exercise its licensing option will be a major catalyst. Assuming the option is exercised, their global development strategy, including potential trials in new indications and combinations, will shape the drug's long-term future.
In conclusion, SHR-A1904 stands as a well-positioned, scientifically robust, and clinically promising antibody-drug conjugate. Backed by the dual strengths of Jiangsu Hengrui's domestic focus and Merck KGaA's global reach, it has a clear path forward. Its ultimate success will hinge on the execution of its ambitious clinical program and its ability to deliver a definitively superior therapeutic benefit in a highly competitive but clinically important new arena of oncology.
Published at: September 14, 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.