
95% of researchers rate our articles as excellent or good
Learn more about the work of our research integrity team to safeguard the quality of each article we publish.
Find out more
STUDY PROTOCOL article
Front. Immunol. , 25 February 2025
Sec. Cancer Immunity and Immunotherapy
Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1519545
This article is part of the Research Topic Checkpoint Immunotherapy: Reshaping the Landscape of Gastrointestinal Cancer Treatment View all 9 articles
Background: Gastric cancer (GC) is one of the most prevalent malignant tumors worldwide, often diagnosed at an advanced stage with a poor prognosis. Paclitaxel, nab-paclitaxel, and irinotecan, either as monotherapies or in combination with ramucirumab, are currently standard second-line treatments for GC. However, the efficacy of these therapies is limited, necessitating the development of new combination strategies to improve response rates. Immune checkpoint inhibitors (ICIs) have shown success in first-line treatment for advanced GC, leading to interest in immune rechallenge strategies for second-line treatment. Re-challenging patients with ICIs after progression on first-line treatment may restore immune responses and provide additional clinical benefit. Recently, cadonilimab (AK104), a bispecific antibody targeting PD-1 and CTLA-4, has demonstrated promising antitumor activity when combined with chemotherapy in advanced gastric and gastroesophageal junction (GEJ) adenocarcinoma. However, the efficacy and safety of nab-paclitaxel combined with AK104 for the treatment of advanced GC remain unclear. Furthermore, identifying predictive biomarkers of efficacy is essential to developing personalized treatment strategies. This study aims to explore the safety and efficacy of nab-paclitaxel combined with AK104 as a second-line treatment for patients who have progressed after first-line chemoimmunotherapy, focusing on evaluating the therapeutic effect of ICIs rechallenge in gastric cancer.
Methods: This is a prospective, multicenter, open-label, single-arm Phase II clinical study. Eligible patients were histologically or cytologically diagnosed with unresectable recurrent or metastatic GC, failed first-line chemotherapy in combination with immune checkpoint inhibitor, aged between 18-75 years old, expected survival ≥3 months, and with a physical status of 0 or 1 in the Eastern Cooperative Cancer Group (ECOG). Enrolled patients will receive intravenous cadonilimab (AK104) 6 mg/kg on days 1, and 15, and intravenous nab-paclitaxel 100 mg/m2 every four weeks on days 1, 8, and 15. The primary endpoints were objective response rate (ORR), and secondary endpoints were disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). The exploratory objective was to identify biomarkers associated with efficacy, mechanism of action, and safety. A total of 59 participants were planned to be recruited using Simon’s two-stage design. The trial was initiated in June 2024 in China.
Discussion: This study is the first prospective trial to evaluate the combination of nab-paclitaxel and cadonilimab as second-line treatment after first-line chemoimmunotherapy failure. By investigating immune rechallenge, it aims to reactivate anti-tumor immune responses and improve clinical outcomes in GC patients. The exploration of predictive biomarkers, such as ctDNA, TMB, MSI, PD-L1 expression, TIL profiles, and gut microbiota, will help personalize treatment and identify patients most likely to benefit from immune rechallenge. This trial could provide valuable insights into overcoming immune resistance and contribute to developing a promising second-line therapeutic strategy for advanced GC.
Clinical trial registration: ClinicalTrials.Gov, identifier NCT06349967
Gastric cancer (GC) is the fifth most common cancer worldwide, with over 2 million new cases and over 611,720 deaths expected in 2024, ranking it as the fourth leading cause of cancer-related deaths (1). Most GC cases are diagnosed at an advanced stage, making radical surgical resection impossible. Currently, fluorouracil-based systemic chemotherapy is the primary treatment for advanced or metastatic GC. However, chemotherapy alone has limited efficacy, and treatment advancements have reached a bottleneck. Recently, immunotherapy has emerged as a promising treatment, showing effective progress in various tumor types (2–4). Immune checkpoint inhibitors (ICIs), such as PD-1 and CTLA-4 inhibitors, restore T cell function by alleviating the immunosuppressive effects of the tumor microenvironment, enabling T cells to effectively attack tumor cells. Studies, including CheckMate 649, ORIENT-16, KEYNOTE-859, GEMSTONE-303, and COMPASSION-15/AK104-302, have demonstrated that combining chemotherapy with ICIs (Nivolumab, Sintilimab, Pembrolizumab, Sugmilimab, and Cadonilimab) significantly improves overall survival (OS) and progression-free survival (PFS) compared to chemotherapy alone, facilitating the transition from first-line treatments to the broader adoption of chemoimmunotherapy in advanced GC (5–10). Despite these improvements, the 5-year survival rate for advanced or metastatic GC remains low (approximately 5-20%), and most patients experience disease progression during immunotherapy. After progression on first-line treatment, current second-line therapies include single-agent chemotherapy (paclitaxel, irinotecan, docetaxel, nab-paclitaxel) or paclitaxel combined with ramucirumab (11–14). However, response rates for these treatments are limited (approximately 10-30%), underscoring the need for new combination strategies to enhance second-line treatment efficacy for GC (15–18).
Tumor cells evade immune detection by upregulating immune checkpoint molecules such as CTLA-4 and PD-1 on T lymphocytes. In gastric adenocarcinoma, high expressions of PD-L1 (around 40%) and CTLA-4 (around 85%) are associated with poor prognosis (19–22). Dual-targeted immunotherapy, validated in both preclinical and clinical studies, has shown efficacy, with CTLA-4 and PD-1 inhibitors working synergistically to restore T cell function through distinct mechanisms (23–27). CTLA-4 upregulation during T cell activation suppresses T cell activity, particularly on tumor-infiltrating regulatory T cells (Tregs) and exhausted effector T cells (Teffs). Blockade of CTLA-4 enhances T cell activation and reduces Treg infiltration, alleviating the immunosuppressive tumor microenvironment. Similarly, PD-1 upregulation following T cell activation suppresses T cell responses by binding its ligand, and inhibiting PD-1/PD-L1 signaling can reinvigorate tumor-reactive T cells. However, recent studies suggest that PD-L1 blockade may paradoxically promote Treg activity, leading to therapeutic resistance, which can be reversed by depleting Tregs (28). Additionally, context-dependent PD-(L)1 checkpoint activation induced by CTLA4-Ig therapy may further suppress T cell activity (29). These findings highlight the potential synergy between checkpoint inhibitors and combination therapies. Chemotherapy also enhances immune responses by inducing tumor apoptosis, upregulating MHC-I, promoting dendritic cell maturation, and inhibiting immunosuppressive cells like Tregs, MDSCs, and TAMs. Combining ICIs with chemotherapy has shown synergistic anti-tumor effects, supporting the rationale for combining chemotherapy with dual immune checkpoint blockade (anti-CTLA-4 and anti-PD-1) in solid tumors (30, 31).
Nab-paclitaxel, a novel formulation of paclitaxel, improves drug concentration and uptake in tumor tissues compared to traditional solvent-based paclitaxel. The ABSOLUTE study showed that nab-paclitaxel is non-inferior to weekly solvent-based paclitaxel and has been recommended as a standard second-line treatment for GC (32). However, its efficacy as a single-agent second-line treatment remains limited, highlighting the need for novel strategies to improve clinical outcomes.
Cadonilimab (AK104) is a bispecific antibody that targets both the PD-1 and CTLA-4 immune checkpoint pathways, utilizing a 4-valent IgG1-ScFv format. It reverses T-cell depletion by facilitating the endocytosis of cell-surface PD-1 and CTLA-4 receptors, which subsequently induces the secretion of IL-2 and IFN-γ. This mechanism not only reduces immune-related adverse events (irAEs) but also enhances the ability of T cells to kill tumor cells (33). Cadonilimab is currently approved in China for treating recurrent or metastatic cervical cancer that has progressed during or after platinum-based chemotherapy (34). In the multicenter, open-label Phase 1b/2 COMPASSION-03 trial, cadonilimab demonstrated significant antitumor activity and a manageable safety profile in patients with advanced solid tumors (9, 23, 35, 36). More recently, in the Phase 3 COMPASSION-15/AK104-302 trial, cadonilimab combined with chemotherapy as a first-line treatment for patients with HER2-negative unresectable advanced or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma showed an objective remission rate (ORR) of up to 65.2% and an overall survival (OS) of up to 15 months (37). Additionally, this combination therapy reduced the risk of death by 44% in patients with high PD-L1 expression (CPS ≥ 5) and by 30% in those with low PD-L1 expression (CPS < 5).
This study aims to evaluate the efficacy and safety of nab-paclitaxel combined with cadonilimab as a second-line treatment for advanced GC following the failure of first-line chemoimmunotherapy (PD1 inhibitors). Additionally, emerging evidence highlights the critical role of gut microbiota and predictive biomarkers in shaping ICI responses, but their integration into GC treatment remains limited. This study will explore diagnostic biomarkers and gut microbiota as predictive and prognostic factors, providing deeper insights into the mechanisms of treatment resistance and therapeutic efficacy. By combining these exploratory analyses with efficacy assessments, the study represents a significant advancement toward personalized immunotherapy, addressing unmet clinical needs in the management of GC.
This prospective, multicenter, open-label, single-arm phase II clinical study aims to evaluate the efficacy and safety of nab-paclitaxel in combination with cadonilimab (AK104) as a second-line treatment for patients with gastric cancer (GC) who have failed first-line fluorouracil-based or platinum-based combination immunotherapy. The study design is illustrated in Figure 1.
Eligible patients are those with histologically or cytologically confirmed unresectable, recurrent, or metastatic GC who have experienced disease progression following first-line chemotherapy combined with immune checkpoint inhibitors (PD-1/PD-L1). Patients enrolled in the study will receive nab-paclitaxel in combination with cadonilimab until either disease progression (PD) or the onset of treatment intolerance.
The study will prospectively collect data on overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and quality of life (QoL), alongside a comprehensive assessment of the medication’s safety profile.
The inclusion and exclusion criteria are detailed in Table 1.
Cadonilimab (AK104): Administered at a dose of 6 mg/kg via intravenous infusion over 30-60 minutes on Days 1 and 15 of each 28-day cycle (q28d).
Nab-paclitaxel: Administered at a dose of 100 mg/m² via intravenous infusion over 30-40 minutes, initiated 30 minutes after the completion of the cadonilimab (AK104) infusion, on Days 1, 8, and 15 of each 28-day cycle (q28d).
Eligible participants will receive nab-paclitaxel in combination with cadonilimab until one of the following occurs: PD, death, loss to follow-up, unacceptable toxicity, withdrawal of informed consent, or other treatment termination criteria as specified in the study protocol.
1. To evaluate the ORR of nab-paclitaxel in combination with cadonilimab (AK104) as a second-line treatment for GC patients who have failed first-line fluorouracil-based or platinum-based combination immunotherapy.
1. To assess the efficacy of nab-paclitaxel combined with cadonilimab (AK104) in the second-line treatment of GC patients who have failed first-line fluorouracil-based or platinum-based combination immunotherapy, including DCR, PFS, and OS.
2. To evaluate the safety, tolerability, and impact on patient QoL of nab-paclitaxel in combination with cadonilimab (AK104).
1. To investigate the antitumor efficacy, mechanism of action, and potential resistance mechanisms of nab-paclitaxel combined with cadonilimab (AK104).
2. To explore the role of tumor tissue PD-L1 expression, blood immune cell subpopulations, and serum cytokine levels as potential predictors of treatment efficacy.
3. To examine the role of liquid biopsy biomarkers, such as circulating tumor DNA (ctDNA), cell-free DNA (cf-DNA), and blood tumor mutational burden (bTMB), as predictors of efficacy, with a focus on their capacity to monitor minimal residual disease or treatment response.
4. To explore the association between gut microbiota composition and immunotherapy response using large-scale metagenomic analysis, aiming to identify microbial signatures that correlate with treatment outcomes.
5. To assess the impact of the treatment protocols on patient QoL using the QLQ-LC13 and EORTC QLQ-C30 questionnaires, with an emphasis on patient-reported outcomes related to physical and emotional well-being.
Objective response rate (ORR): defined as the proportion of subjects whose tumors achieved complete response (CR) or partial response (PR) after treatment. Evaluated by the Blinded Independent Image Review Committee (BIIRC). This serves as the primary indicator of treatment efficacy.
Disease Control Rate (DCR): defined as the proportion of subjects whose tumors achieved CR, PR, or stable disease (SD) after treatment. This reflects broader disease stabilization benefits.
Progression-free survival (PFS): defined as the time from randomization to tumor progression or death due to any cause, providing insights into treatment durability.
Overall survival (OS): defined as the time from randomization to death due to any cause (last follow-up for patients lost to follow-up; end of follow-up date for patients alive at the end of the study). This serves as a critical measure of long-term efficacy.
Accompanying diagnostic biomarkers and gut flora characterization: This exploratory analysis aims to identify predictive and prognostic biomarkers, with a particular focus on gut microbiota and its interaction with immunotherapy. These findings may provide novel insights into the mechanisms underlying treatment responses and immune-related adverse events (irAEs).
Laboratory tests, including hematology, liver and kidney function, electrolytes, cardiac markers, thyroid function, coagulation, and transmission nine tests, were performed on the first day of each treatment cycle. Tumor biomarker assessments, including carbohydrate antigen 19-9 (CA19-9), CA125, carcinoembryonic antigen (CEA), and alpha-fetoprotein (AFP), were also conducted. Additionally, 12-lead electrocardiograms and echocardiograms were performed at the same time. These tests were performed according to the protocol, with additional assessments performed if clinically indicated.
Antitumor efficacy was evaluated every 8 weeks (approximately at the end of every two dosing cycles) via CT or MRI examinations (chest, abdomen, pelvis, and any other site suspected of having tumor lesions). Tumor imaging response was assessed according to RECIST version 1.1. Tumor assessments may be conducted more frequently if clinically necessary, based on the investigator’s judgment.
All procedures were conducted in accordance with the study protocol and with informed consent from the participants.
Safety assessments will be conducted throughout the study, with participants closely monitored for adverse events (AEs) until resolution, stabilization, or confirmation of non-clinical significance.
AE Definition: defined as any untoward medical occurrence in a participant receiving the investigational product, irrespective of its causal relationship to the treatment. AEs will be evaluated for severity (graded per NCI-CTCAE v5.0), duration, and relationship to the study treatment.
Serious Adverse Event (SAE) Definition: defined as any event that results in death, is life-threatening, causes significant disability, requires hospitalization or prolongs an existing hospitalization, leads to congenital anomalies, or constitutes other medically significant conditions. SAEs must be reported to the regulatory authority, ethics committee, and sponsor within 24 hours, with follow-up reports submitted as needed. All AEs/SAEs must be thoroughly documented in the case report form (CRF), including details on onset, duration, resolution, and any interventions taken.
The study team will follow the study protocol and standard operating procedures (SOPs) for AE/SAE management, implementing necessary measures such as dose adjustments or treatment discontinuation when required.
For low-grade adverse events (Grade 1 or 2), symptomatic and topical treatments are recommended. Persistent low-grade events or severe events (Grade ≥3) should be managed with systemic corticosteroids, such as prednisone or intravenous equivalents.
Discontinuation of AK104 therapy is not mandatory for Grade 3 or 4 inflammatory responses (e.g., inflammation at metastatic sites or lymph nodes) attributable to a localized tumor response. In cases of multiple concurrent low-grade AEs that individually would not necessitate therapy termination, the decision to discontinue AK104 treatment will be at the investigator’s discretion.
The investigator will evaluate the severity of immune-related adverse events (irAEs) using the NCI CTCAE version 5.0 grading criteria and adjust AK104 therapy as necessary. General management recommendations for irAEs are outlined in Table 2.
Overall survival (OS) analysis will be conducted throughout the trial period. During the follow-up phase, subjects will be assessed every three months to document their survival status, including PD, AEs, and QoL where applicable. Follow-up visits may be conducted through in-person consultations, phone calls, or electronic surveys, depending on patient circumstances.
Follow-up will continue until the subject’s death, loss to follow-up, or the end of the study period. All data collected during follow-up will be systematically recorded in the study database. Measures will be implemented to ensure adherence to follow-up schedules, including regular reminders and patient support initiatives.
This study focuses on biomarker analysis and gut microbiota characterization to explore the predictive value of peripheral biomarkers in assessing disease activity and survival benefits associated with AK104 treatment, leveraging advancements in liquid biopsy technologies and their therapeutic potential.
Peripheral blood samples were collected before the first dose (Day 1, Cycle 1) and at disease progression. Fresh whole blood was processed within 2 hours to extract plasma, which was stored at -80°C for next-generation sequencing (NGS) analysis. NGS was used to evaluate cancer-related genes, including ctDNA, cfDNA somatic mutations, and bTMB. Changes in immune cell subsets, cytokines, and tumor immunotherapy biomarkers were also assessed.
Fecal samples, collected before treatment initiation, were analyzed using large-scale metagenomic sequencing. This approach provided comprehensive taxonomic and functional profiling to assess associations between gut microbiota composition and immunotherapy response.
Data from biomarker and microbiota analyses were evaluated using multivariate regression and survival models, with adjustments for confounding factors. All patients provided informed consent, and the study protocol received approval from the institutional review board (IRB).
Sample size calculations were performed using Simon’s optimal two-stage design to balance ethical considerations and resource efficiency. This design minimizes the expected sample size under the null hypothesis while ensuring adequate power to detect a meaningful treatment effect if the alternative hypothesis is true. Simon’s two-stage design was chosen due to its widespread use in early-phase clinical trials to assess efficacy and safety efficiently.
The minimax design parameters were set as follows: an alpha error of 5% and a power (1-β) of 80% were used for calculations. The minimax two-stage design resulted in parameters (6/31, 15/53). Stage 1: Enroll 31 patients. If ≤6 patients achieve ORR, the trial is terminated due to futility. Stage 2: If >6 responses are observed, an additional 22 patients are enrolled, resulting in a total of 53 patients. Success Criteria: The treatment regimen is considered successful if ≥15 patients achieve ORR. To account for potential patient dropout, the sample size was increased by 10%, resulting in a final planned enrollment of 59 patients. PFS and OS will be analyzed using the Kaplan-Meier method. Median survival times and 95% confidence intervals (CI) will be reported. Differences between groups will be assessed using the log-rank test. If applicable, Cox proportional hazards regression analysis will be performed to evaluate the impact of baseline covariates on survival outcomes. ORR, DCR, and AEs will be analyzed using descriptive statistics. The Clopper-Pearson method will be used to calculate 95% confidence intervals for proportions. Subgroup analyses may be conducted based on predefined stratification factors. Sample size calculations were performed using PASS 2024 software (version 24.0.2, NCSS).
Exploratory analyses will be conducted to identify predictive and prognostic biomarkers using advanced immunoassays and bioinformatics tools. Adverse events will be graded according to CTCAE v5.0, and comparisons between different severity grades will be performed using chi-square or Fisher’s exact tests. This statistical approach ensures rigorous evaluation of efficacy and safety while accommodating the exploratory nature of biomarker identification.
Although first-line chemotherapy regimens for advanced gastric cancer (GC) have improved, the survival times remains below 12 months, highlighting the need for further efficacy advancements. Immunotherapy has made significant strides in treating advanced GC, overcoming the long-standing survival limitations associated with traditional chemotherapy (38–40). Notably, the CheckMate-649 trial, a randomized, open-label, multicenter Phase III study, evaluated nivolumab combined with chemotherapy, nivolumab plus ipilimumab, and single-agent chemotherapy in HER2-negative advanced GC patients. Results showed that nivolumab plus chemotherapy significantly improved OS and PFS compared to chemotherapy alone, with an acceptable safety profile (41). Importantly, in patients with PD-L1 CPS ≥ 5, nivolumab in combination with chemotherapy reduced the risk of death by 29% compared to chemotherapy alone. As a result, nivolumab combined with chemotherapy has become a recommended first-line treatment option in clinical guidelines from CSCO, NCCN, and ESMO. Similarly, the ATTRACTION-04 trial, a randomized multicenter Phase II/III study, demonstrated that first-line nivolumab with chemotherapy significantly improved PFS in Asian patients with HER2-negative advanced or recurrent GC (PFS: 10-45 months vs. 8-34 months; HR 0.68; 98.51% CI: 0.51-0.90; P=0.0007), suggesting it may become the new standard of care (42). Additional studies, including ORIENT-16, RATIONALE-305, KEYNOTE-859, GEMSTONE-303, and COMPASSION-15/AK104-302, showed that combining chemotherapy with PD-1/PD-L1 monoclonal antibodies such as sindilizumab, tirilizumab, pabolizumab, sugemalimab, and cadonilimab improved median overall survival (mOS) to 13-15 months. In patients with high PD-L1 expression, mOS reached 15-18 months, with an ORR of approximately 60%.
The combination of ICIs and chemotherapy has now become the standard first-line treatment for advanced GC without driver gene mutations, including in unresectable locally advanced GC and perioperative settings (43–45). Despite these significant improvements in prognosis, around 40% of patients still experience PD, and OS often falls short of expectations. Re-challenge with ICIs, which involves reintroducing these agents after PD or serious irAEs, has been extensively studied in melanoma and urologic cancers (46–48). ICI rechallenge works by either reactivating the normal immune cycle or bypassing immune dysfunction to resensitize tumor cells to ICIs (49–51). Tumor cells that initially responded to ICIs may still harbor susceptible populations, and the interactions between the immune system and the tumor microenvironment may provide new opportunities for anti-tumor responses. Additionally, immunotherapy can alter the tumor neoantigen profile, enabling tumors to evade recognition by memory T cells, but ICI rechallenge can restore T cells’ ability to recognize these neoantigens (4, 52). Studies such as CheckMate 066/067 and KEYNOTE-010 have demonstrated that rechallenge with nivolumab and pembrolizumab resulted in lesion shrinkage and significant OS improvements in patients with advanced melanoma and PD-L1-positive advanced non-small-cell lung cancer (NSCLC), with no additional AEs reported (53–56).
Although chemotherapy combined with immunotherapy demonstrates synergistic effects through the promotion of immunogenic cell death (ICD) and the release of neoantigens, the rechallenge of ICIs in advanced GC remains underexplored due to limited clinical evidence and the lack of large-scale prospective studies. Furthermore, identifying biomarkers for ICI rechallenge in advanced GC is crucial for facilitating precise, individualized immunotherapy and improving the effectiveness of combination therapies. Significant progress has been made in identifying biomarkers for gastrointestinal cancers, with PD-L1 expression, microsatellite instability (MSI), and mismatch repair deficiency (dMMR) emerging as key indicators (57). Ongoing research is also focusing on other biomarkers, including tumor mutational burden (TMB), circulating tumor DNA (ctDNA), Epstein-Barr virus (EBV), and the gut microbiome. Advanced immunoassays, multi-omics approaches, and bioinformatics tools offer additional potential for identifying biomarkers predictive of positive responses to ICI rechallenge. By integrating these biomarkers with clinicopathological features, we can more effectively stratify advanced GC patients undergoing ICI treatment, explore mechanisms of ICI drug resistance, identify potential strategies for sensitization, and ultimately improving overall prognosis (58–62).
Managing irAEs is a critical aspect of immunotherapy. As a bispecific antibody targeting both PD-1 and CTLA-4, cadonilimab may have a unique irAE profile. Common irAEs include dermatologic, gastrointestinal, hepatic, and endocrine disorders, all of which require timely diagnosis and management. Strategies such as early corticosteroid intervention, immune-modulating agents, and careful monitoring are essential to mitigate irAEs. Our study emphasizes the need for standardized irAE management protocols, especially for novel agents like cadonilimab.
This study has several strengths, including the investigation of a novel combination therapy, exploration of predictive biomarkers, and well-defined patient stratification. However, it also has limitations, as it was conducted within a single geographic region and focused primarily on an Asian population. While the findings offer valuable insights into the efficacy of cadonilimab in this group, their generalizability to other populations remains uncertain. Gastric cancer epidemiology, genetic diversity, and healthcare access vary globally, which may influence treatment responses and outcomes. Additionally, potential confounding from prior immunotherapy treatment could impact the interpretation of results. Future multicenter and global studies are needed to validate these results and ensure the broader applicability of cadonilimab-based regimens.
This Phase II clinical trial is the first to evaluate nab-paclitaxel combined with cadonilimab (AK104) as a second-line treatment for advanced GC. The study aims to investigate the efficacy and safety of ICI rechallenge in patients who have progressed after first-line chemoimmunotherapy, focusing on immune resistance mechanisms and predictive biomarkers of susceptibility. By exploring real-world rechallenge strategies, this research provides evidence-based insights to optimize immunotherapy outcomes and paves the way for future studies, highlighting cadonilimab’s potential to transform treatment for advanced gastric cancer.
The studies involving humans were approved by Biomedical Ethics Review Committee, West China Hospital, Sichuan University. The studies were conducted in accordance with the local legislation and institutional requirements. The participants provided their written informed consent to participate in this study.
JW: Data curation, Software, Writing – original draft. QH: Methodology, Supervision, Writing – review & editing. XC: Supervision, Writing – review & editing. CS: Supervision, Writing – review & editing. ZC: Supervision, Writing – review & editing. WZ: Supervision, Writing – review & editing. YY: Supervision, Writing – review & editing. BZ: Supervision, Writing – review & editing. HG: Supervision, Writing – review & editing. KY: Supervision, Writing – review & editing. FB: Conceptualization, Supervision, Writing – review & editing. PZ: Methodology, Validation, Writing – review & editing. ML: Conceptualization, Methodology, Project administration, Writing – review & editing.
The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by 1.3.5 Project for Disciplines of Excellence, West China Hospital, Si-chuan University (Grant No. ZYJC21043). Science and technology innovation talent project of Sichuan Science and Technology Department (2020JDRC0025). Sichuan Provincial Science and Technology Department 2023 key research and development project in the field of social development science and technology (2023YFS0111).
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
The author(s) declare that no Generative AI was used in the creation of this manuscript.
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
1. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA: Cancer J Clin. (2024) 74:12–49. doi: 10.3322/caac.21820
2. Alsina M, Arrazubi V, Diez M, Tabernero J. Current developments in gastric cancer: from molecular profiling to treatment strategy. Nat Rev Gastroenterol hepatology. (2023) 20:155–70. doi: 10.1038/s41575-022-00703-w
3. Svrcek M, Voron T, André T, Smyth EC, de la Fouchardière C. Improving individualised therapies in localised gastro-oesophageal adenocarcinoma. Lancet Oncol. (2024) 25:e452–e63. doi: 10.1016/s1470-2045(24)00180-3
4. Patel MA, Kratz JD, Lubner SJ, Loconte NK, Uboha NV. Esophagogastric cancers: integrating immunotherapy therapy into current practice. J Clin oncology: Off J Am Soc Clin Oncol. (2022) 40:2751–62. doi: 10.1200/jco.21.02500
5. Janjigian YY, Ajani JA, Moehler M, Shen L, Garrido M, Gallardo C, et al. First-line nivolumab plus chemotherapy for advanced gastric, gastroesophageal junction, and esophageal adenocarcinoma: 3-year follow-up of the phase iii checkmate 649 trial. J Clin oncology: Off J Am Soc Clin Oncol. (2024) 42:2012–20. doi: 10.1200/jco.23.01601
6. Xu J, Jiang H, Pan Y, Gu K, Cang S, Han L, et al. Sintilimab plus chemotherapy for unresectable gastric or gastroesophageal junction cancer: the orient-16 randomized clinical trial. Jama. (2023) 330:2064–74. doi: 10.1001/jama.2023.19918
7. Rha SY, Oh DY, Yañez P, Bai Y, Ryu MH, Lee J, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for her2-negative advanced gastric cancer (Keynote-859): A multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. (2023) 24:1181–95. doi: 10.1016/s1470-2045(23)00515-6
8. Zhang X, Wang J, Wang G, Zhang Y, Fan Q, Chuangxin L, et al. Lba79 gemstone-303: prespecified progression-free survival (Pfs) and overall survival (Os) final analyses of a phase iii study of sugemalimab plus chemotherapy vs placebo plus chemotherapy in treatment-na&Xef;Ve advanced gastric or gastroesophageal junction (G/gej) adenocarcinoma. Ann Oncol. (2023) 34:S1319. doi: 10.1016/j.annonc.2023.10.080
9. Gao X, Ji K, Jia Y, Shan F, Chen Y, Xu N, et al. Cadonilimab with chemotherapy in her2-negative gastric or gastroesophageal junction adenocarcinoma: the phase 1b/2 compassion-04 trial. Nat Med. (2024) 30:1943–51. doi: 10.1038/s41591-024-03007-5
10. A Randomized, Double-Blind, Multicenter, Phase Iii Study Comparing the Efficacy and Safety of Ak104 Plus Oxaliplatin and Capecitabine (Xelox) Versus Placebo Plus Xelox as First-Line Treatment for Locally Advanced Unresectable or G/Gej Adenocarcinoma (2021). Available online at: https://clinicaltrials.gov/study/NCT05008783.
11. Ward ZJ, Gaba Q, Atun R. Cancer incidence and survival for 11 cancers in the commonwealth: A simulation-based modelling study. Lancet Oncol. (2024) 25:1127–34. doi: 10.1016/s1470-2045(24)00336-x
12. Pavlakis N, Shitara K, Sjoquist K, Martin A, Jaworski A, Tebbutt N, et al. Integrate iia phase iii study: regorafenib for refractory advanced gastric cancer. J Clin oncology: Off J Am Soc Clin Oncol. (2024) 43(4):453–63. doi: 10.1200/jco.24.00055
13. Kang YK, Kim HD, Yook JH, Park YK, Lee JS, Kim YW, et al. Neoadjuvant docetaxel, oxaliplatin, and S-1 plus surgery and adjuvant S-1 for resectable advanced gastric cancer: updated overall survival outcomes from phase iii prodigy. J Clin oncology: Off J Am Soc Clin Oncol. (2024) 42:2961–65. doi: 10.1200/jco.23.02167
14. Hegewisch-Becker S, Mendez G, Chao J, Nemecek R, Feeney K, Van Cutsem E, et al. First-line nivolumab and relatlimab plus chemotherapy for gastric or gastroesophageal junction adenocarcinoma: the phase ii relativity-060 study. J Clin oncology: Off J Am Soc Clin Oncol. (2024) 42:2080–93. doi: 10.1200/jco.23.01636
15. Tougeron D, Dahan L, Evesque L, Le Malicot K, El Hajbi F, Aparicio T, et al. Folfiri plus durvalumab with or without tremelimumab in second-line treatment of advanced gastric or gastroesophageal junction adenocarcinoma: the prodige 59-ffcd 1707-durigast randomized clinical trial. JAMA Oncol. (2024) 10:709–17. doi: 10.1001/jamaoncol.2024.0207
16. Kim CG, Jung M, Kim HS, Lee CK, Jeung HC, Koo DH, et al. Trastuzumab combined with ramucirumab and paclitaxel in patients with previously treated human epidermal growth factor receptor 2-positive advanced gastric or gastroesophageal junction cancer. J Clin oncology: Off J Am Soc Clin Oncol. (2023) 41:4394–405. doi: 10.1200/jco.22.02122
17. Wang F, Shen L, Guo W, Liu T, Li J, Qin S, et al. Fruquintinib plus paclitaxel versus placebo plus paclitaxel for gastric or gastroesophageal junction adenocarcinoma: the randomized phase 3 frutiga trial. Nat Med. (2024) 30:2189–98. doi: 10.1038/s41591-024-02989-6
18. Xu RH, Zhang Y, Pan H, Feng J, Zhang T, Liu T, et al. Efficacy and safety of weekly paclitaxel with or without ramucirumab as second-line therapy for the treatment of advanced gastric or gastroesophageal junction adenocarcinoma (Rainbow-asia): A randomised, multicentre, double-blind, phase 3 trial. Lancet Gastroenterol hepatology. (2021) 6:1015–24. doi: 10.1016/s2468-1253(21)00313-7
19. Chen Y, Jia K, Chong X, Xie Y, Jiang L, Peng H, et al. Implications of pd-L1 expression on the immune microenvironment in her2-positive gastric cancer. Mol cancer. (2024) 23:169. doi: 10.1186/s12943-024-02085-w
20. Cousin S, Guégan JP, Shitara K, Palmieri LJ, Metges JP, Pernot S, et al. Identification of microenvironment features associated with primary resistance to anti-pd-1/pd-L1 + Antiangiogenesis in gastric cancer through spatial transcriptomics and plasma proteomics. Mol cancer. (2024) 23:197. doi: 10.1186/s12943-024-02092-x
21. Weng N, Zhou C, Zhou Y, Zhong Y, Jia Z, Rao X, et al. Ikzf4/nono-rab11fip3 axis promotes immune evasion in gastric cancer via facilitating pd-L1 endosome recycling. Cancer letters. (2024) 584:216618. doi: 10.1016/j.canlet.2024.216618
22. Dovedi SJ, Elder MJ, Yang C, Sitnikova SI, Irving L, Hansen A, et al. Design and efficacy of a monovalent bispecific pd-1/ctla4 antibody that enhances ctla4 blockade on pd-1(+) activated T cells. Cancer Discovery. (2021) 11:1100–17. doi: 10.1158/2159-8290.Cd-20-1445
24. Principe N, Phung AL, Stevens KLP, Elaskalani O, Wylie B, Tilsed CM, et al. Anti-metabolite chemotherapy increases lag-3 expressing tumor-infiltrating lymphocytes which can be targeted by combination immune checkpoint blockade. J Immunother Cancer. (2024) 12. doi: 10.1136/jitc-2023-008568
25. Jiang Y, Bei W, Li W, Huang Y, He S, Zhu X, et al. Single-cell transcriptome analysis reveals evolving tumour microenvironment induced by immunochemotherapy in nasopharyngeal carcinoma. Clin Trans Med. (2024) 14:e70061. doi: 10.1002/ctm2.70061
26. Skoulidis F, Araujo HA, Do MT, Qian Y, Sun X, Cobo AG, et al. Ctla4 blockade abrogates keap1/stk11-related resistance to pd-(L)1 inhibitors. Nature. (2024) 635(8038):462–71. doi: 10.1038/s41586-024-07943-7
27. Wang K, Coutifaris P, Brocks D, Wang G, Azar T, Solis S, et al. Combination anti-pd-1 and anti-ctla-4 therapy generates waves of clonal responses that include progenitor-exhausted cd8(+) T cells. Cancer Cell. (2024) 42:1582–97.e10. doi: 10.1016/j.ccell.2024.08.007
28. van Gulijk M, van Krimpen A, Schetters S, Eterman M, van Elsas M, Mankor J, et al. Pd-L1 checkpoint blockade promotes regulatory T cell activity that underlies therapy resistance. Sci Immunol. (2023) 8:eabn6173. doi: 10.1126/sciimmunol.abn6173
29. Oxley EP, Kershaw NJ, Louis C, Goodall KJ, Garwood MM, Jee Ho SM, et al. Context-restricted pd-(L)1 checkpoint agonism by ctla4-ig therapies inhibits T cell activity. Cell Rep. (2024) 43:114834. doi: 10.1016/j.celrep.2024.114834
30. Chen YY, Zeng XT, Gong ZC, Zhang MM, Wang KQ, Tang YP, et al. Euphorbia pekinensis rupr. Sensitizes colorectal cancer to pd-1 blockade by remodeling the tumor microenvironment and enhancing peripheral immunity. Phytomedicine: Int J phytotherapy phytopharmacology. (2024) 135:156107. doi: 10.1016/j.phymed.2024.156107
31. Skubleny D, Purich K, McLean DR, Martins-Filho SN, Buttenschoen K, Haase E, et al. The tumour immune microenvironment drives survival outcomes and therapeutic response in an integrated molecular analysis of gastric adenocarcinoma. Clin Cancer research: an Off J Am Assoc Cancer Res. (2024) 30(23):5385–98. doi: 10.1158/1078-0432.Ccr-23-3523
32. Shitara K, Takashima A, Fujitani K, Koeda K, Hara H, Nakayama N, et al. Nab-paclitaxel versus solvent-based paclitaxel in patients with previously treated advanced gastric cancer (Absolute): an open-label, randomised, non-inferiority, phase 3 trial. Lancet Gastroenterol hepatology. (2017) 2:277–87. doi: 10.1016/s2468-1253(16)30219-9
33. Pang X, Huang Z, Zhong T, Zhang P, Wang ZM, Xia M, et al. Cadonilimab, a tetravalent pd-1/ctla-4 bispecific antibody with trans-binding and enhanced target binding avidity. mAbs. (2023) 15:2180794. doi: 10.1080/19420862.2023.2180794
34. Lou H, Cai H, Huang X, Li G, Wang L, Liu F, et al. Cadonilimab combined with chemotherapy with or without bevacizumab as first-line treatment in recurrent or metastatic cervical cancer (Compassion-13): A phase 2 study. Clin Cancer research: an Off J Am Assoc Cancer Res. (2024) 30:1501–08. doi: 10.1158/1078-0432.Ccr-23-3162
35. Frentzas S, Gan HK, Cosman R, Coward J, Tran B, Millward M, et al. A phase 1a/1b first-in-human study (Compassion-01) evaluating cadonilimab in patients with advanced solid tumors. Cell Rep Med. (2023) 4:101242. doi: 10.1016/j.xcrm.2023.101242
36. Gao X, Xu N, Li Z, Shen L, Ji K, Zheng Z, et al. Safety and antitumour activity of cadonilimab, an anti-pd-1/ctla-4 bispecific antibody, for patients with advanced solid tumours (Compassion-03): A multicentre, open-label, phase 1b/2 trial. Lancet Oncol. (2023) 24:1134–46. doi: 10.1016/s1470-2045(23)00411-4
38. Rached L, Laparra A, Sakkal M, Danlos FX, Barlesi F, Carbonnel F, et al. Toxicity of immunotherapy combinations with chemotherapy across tumor indications: current knowledge and practical recommendations. Cancer Treat Rev. (2024) 127:102751. doi: 10.1016/j.ctrv.2024.102751
39. Shah MA, Kennedy EB, Alarcon-Rozas AE, Alcindor T, Bartley AN, Malowany AB, et al. Immunotherapy and targeted therapy for advanced gastroesophageal cancer: asco guideline. J Clin oncology: Off J Am Soc Clin Oncol. (2023) 41:1470–91. doi: 10.1200/jco.22.02331
40. Liang H, Yan X, Li Z, Chen X, Qiu Y, Li F, et al. Clinical outcomes of conversion surgery following immune checkpoint inhibitors and chemotherapy in stage iv gastric cancer. Int J Surg (London England). (2023) 109:4162–72. doi: 10.1097/js9.0000000000000738
41. Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (Checkmate 649): A randomised, open-label, phase 3 trial. Lancet (London England). (2021) 398:27–40. doi: 10.1016/s0140-6736(21)00797-2
42. Kang YK, Chen LT, Ryu MH, Oh DY, Oh SC, Chung HC, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with her2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (Attraction-4): A randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. (2022) 23:234–47. doi: 10.1016/s1470-2045(21)00692-6
43. Schutte T, Derks S, van Laarhoven HWM. Pembrolizumab plus chemotherapy for advanced gastric cancer. Lancet Oncol. (2024) 25:e51. doi: 10.1016/s1470-2045(23)00621-6
44. Kang YK, Terashima M, Kim YW, Boku N, Chung HC, Chen JS, et al. Adjuvant nivolumab plus chemotherapy versus placebo plus chemotherapy for stage iii gastric or gastro-oesophageal junction cancer after gastrectomy with D2 or more extensive lymph-node dissection (Attraction-5): A randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Gastroenterol hepatology. (2024) 9:705–17. doi: 10.1016/s2468-1253(24)00156-0
45. An M, Mehta A, Min BH, Heo YJ, Wright SJ, Parikh M, et al. Early immune remodeling steers clinical response to first-line chemoimmunotherapy in advanced gastric cancer. Cancer Discovery. (2024) 14:766–85. doi: 10.1158/2159-8290.Cd-23-0857
46. Topp BG, Channavazzala M, Mayawala K, De Alwis DP, Rubin E, Snyder A, et al. Tumor dynamics in patients with solid tumors treated with pembrolizumab beyond disease progression. Cancer Cell. (2023) 41:1680–88.e2. doi: 10.1016/j.ccell.2023.08.004
47. Yochum ZA, Braun DA. Rechallenging with anti-pd-1 therapy in advanced renal cell carcinoma. Lancet (London England). (2024) 404:1280–82. doi: 10.1016/s0140-6736(24)01866-x
48. Sun L, Cohen RB, D’Avella CA, Singh AP, Schoenfeld JD, Hanna GJ. Overall survival, treatment duration, and rechallenge outcomes with ici therapy for recurrent or metastatic hnscc. JAMA network Open. (2024) 7:e2428526. doi: 10.1001/jamanetworkopen.2024.28526
49. Baessler A, Fuchs B, Perkins B, Richens AW, Novis CL, Harrison-Chau M, et al. Tet2 deletion in cd4+ T cells disrupts th1 lineage commitment in memory cells and enhances T follicular helper cell recall responses to viral rechallenge. Proc Natl Acad Sci United States America. (2023) 120:e2218324120. doi: 10.1073/pnas.2218324120
50. Gang X, Yan J, Li X, Shi S, Xu L, Liu R, et al. Immune checkpoint inhibitors rechallenge in non-small cell lung cancer: current evidence and future directions. Cancer letters. (2024) 604:217241. doi: 10.1016/j.canlet.2024.217241
51. Virassamy B, Caramia F, Savas P, Sant S, Wang J, Christo SN, et al. Intratumoral cd8(+) T cells with a tissue-resident memory phenotype mediate local immunity and immune checkpoint responses in breast cancer. Cancer Cell. (2023) 41:585–601.e8. doi: 10.1016/j.ccell.2023.01.004
52. Kuhn NF, Lopez AV, Li X, Cai W, Daniyan AF, Brentjens RJ. Cd103(+) cdc1 and endogenous cd8(+) T cells are necessary for improved cd40l-overexpressing car T cell antitumor function. Nat Commun. (2020) 11:6171. doi: 10.1038/s41467-020-19833-3
53. Robert C, Long GV, Brady B, Dutriaux C, Di Giacomo AM, Mortier L, et al. Five-year outcomes with nivolumab in patients with wild-type braf advanced melanoma. J Clin oncology: Off J Am Soc Clin Oncol. (2020) 38:3937–46. doi: 10.1200/jco.20.00995
54. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, et al. Long-term outcomes with nivolumab plus ipilimumab or nivolumab alone versus ipilimumab in patients with advanced melanoma. J Clin oncology: Off J Am Soc Clin Oncol. (2022) 40:127–37. doi: 10.1200/jco.21.02229
55. Wolchok JD, Chiarion-Sileni V, Rutkowski P, Cowey CL, SChadendorf D, Wagstaff J, et al. Final, 10-year outcomes with nivolumab plus ipilimumab in advanced melanoma. New Engl J Med. (2024) 392(1):11–22. doi: 10.1056/NEJMoa2407417
56. Herbst RS, Garon EB, Kim DW, Cho BC, Perez-Gracia JL, Han JY, et al. Long-term outcomes and retreatment among patients with previously treated, programmed death-ligand 1−Positive, advanced non−Small-cell lung cancer in the keynote-010 study. J Clin oncology: Off J Am Soc Clin Oncol. (2020) 38:1580–90. doi: 10.1200/jco.19.02446
57. Lee CK, Kim HS, Jung M, Kim H, Bae WK, Koo DH, et al. Open-label, multicenter, randomized, biomarker-integrated umbrella trial for second-line treatment of advanced gastric cancer: K-umbrella gastric cancer study. J Clin oncology: Off J Am Soc Clin Oncol. (2024) 42:348–57. doi: 10.1200/jco.23.00971
58. Singh H, Lowder KE, Kapner K, Kelly RJ, Zheng H, McCleary NJ, et al. Clinical outcomes and ctdna correlates for capox betr: A phase ii trial of capecitabine, oxaliplatin, bevacizumab, trastuzumab in previously untreated advanced her2+ Gastroesophageal adenocarcinoma. Nat Commun. (2024) 15:6833. doi: 10.1038/s41467-024-51271-3
59. Shi M, Zeng D, Luo H, Xiao J, Li Y, Yuan X, et al. Tumor microenvironment rna test to predict immunotherapy outcomes in advanced gastric cancer: the times001 trial. Med (New York NY). (2024) 5(11):1378–92. doi: 10.1016/j.medj.2024.07.006
60. Yagisawa M, Taniguchi H, Satoh T, Kadowaki S, Sunakawa Y, Nishina T, et al. Trastuzumab deruxtecan in advanced solid tumors with human epidermal growth factor receptor 2 amplification identified by plasma cell-free DNA testing: A multicenter, single-arm, phase ii basket trial. J Clin oncology: Off J Am Soc Clin Oncol. (2024) 42(32):3817–25. doi: 10.1200/jco.23.02626
61. Bos J, Groen-van Schooten TS, Brugman CP, Jamaludin FS, van Laarhoven HWM, Derks S. The tumor immune composition of mismatch repair deficient and epstein-barr virus-positive gastric cancer: A systematic review. Cancer Treat Rev. (2024) 127:102737. doi: 10.1016/j.ctrv.2024.102737
Keywords: gastric cancer, cadonilimab (AK104), nab-paclitaxel, immunotherapy, phase II clinical trial
Citation: Wei J, Zhang P, Hu Q, Cheng X, Shen C, Chen Z, Zhuang W, Yin Y, Zhang B, Gou H, Yang K, Bi F and Liu M (2025) Nab-paclitaxel combined with cadonilimab (AK104) as second-line treatment for advanced gastric cancer: protocol for a phase II prospective, multicenter, single-arm clinical trial. Front. Immunol. 16:1519545. doi: 10.3389/fimmu.2025.1519545
Received: 30 October 2024; Accepted: 31 January 2025;
Published: 25 February 2025.
Edited by:
Stavros P. Papadakos, Laiko General Hospital of Athens, GreeceReviewed by:
Xing Xiao, Sun Yat-sen University, ChinaCopyright © 2025 Wei, Zhang, Hu, Cheng, Shen, Chen, Zhuang, Yin, Zhang, Gou, Yang, Bi and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Ming Liu, bGl1bWluZzYyOUB3Y2hzY3UuY24=
†These authors have contributed equally to this work and share first authorship
Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.
Research integrity at Frontiers
Learn more about the work of our research integrity team to safeguard the quality of each article we publish.