- Research
- Open access
- Published:
Unveiling the therapeutic potential of HZQYF: exploring the inhibitory impact of a clinical herbal formula on gastric cancer through network pharmacology and transcript analysis
BMC Complementary Medicine and Therapies volume 25, Article number: 142 (2025)
Abstract
Hezi Qingyou Formula (HZQYF) is a clinical formulation known for its efficacy in treating gastrointestinal diseases. Nevertheless, its specific impact and underlying mechanism of action in gastric cancer remain to be fully elucidated. The major components of the formula were precisely identified and characterized using ultra-high-performance liquid chromatography coupled with a tandem mass spectrometer (UHPLC-MS/MS). Network pharmacology and transcript analysis were utilized to identify the targets associated with drug-disease interactions. Subsequently, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome analyses were conducted to unravel the pivotal pathways involved. Furthermore, in vitro experiments were performed to validate the anti-gastric cancer activity of HZQYF, including assessments of cell viability and clonogenic potential. These results revealed that 260 co-expressed targets were identified as shared between HZQYF and gastric cancer. These genes were significantly enriched in biological processes and pathways related to steroid metabolism, gamma-aminobutyric acid (GABA)-A receptor complex, steroid binding activity, extracellular ligand-gated ion channel activity, chemical carcinogenesis-reactive oxygen species, and GABAergic synapse. Furthermore, the principal components of the formula were characterized. Subsequent cell experiments confirmed the formula's ability to inhibit gastric cancer activity and suppress colony formation in vitro. In conclusion, these findings suggest that Hezi Qingyou Formula may exert its anti-gastric cancer activity by influencing reactive oxygen species and modulating GABAergic synapses in-silico methods. This study provides a foundation for further exploration of HZQYF as a potential therapeutic agent for gastric cancer.
Introduction
Gastric cancer (GC) is a prevalent global health concern, ranking as the fifth most common malignancy and the third leading cause of cancer-related deaths worldwide [1]. Treatment options for this condition encompass surgical interventions, radiotherapy, chemotherapy, and targeted therapy [2,3,4,5,6,7,8]. Unfortunately, the clinical treatments involving radiotherapy and chemotherapy often come with significant adverse effects due to the disease's tendency to be diagnosed in advanced stages. Traditional Chinese medicine (TCM), known for its holistic approach and evidence-based therapies, offers promise in improving patient symptoms and mitigating the occurrence of adverse reactions [9,10,11].
Prescriptions in TCM are typically derived from the clinical practical application of TCM, guided by TCM theory. This involves consolidating individual medication experiences in clinical practice and progressively elucidating the appropriate population, dosage, therapeutic characteristics, and clinical benefits. The culmination of this process is to develop a standardized prescription, which can evolve into a new TCM suitable for a specific group of patients. This patient-centered approach focuses on clinical value, reflecting the distinctive role of TCM. It makes the best of the clinical advantages of TCM and is executed through various methods, such as integrating diseases and evidence, focusing on particular diseases and medications, or evidence-based TCM. These strategies clarify the clinical benefits for patients, thereby ensuring the efficacy of the treatment.
Based on the clinical application of TCM theory, we have identified and formulated a clinical formula called Hezi Qingyou Formula (HZQYF) to treat gastrointestinal disorders. Our research has demonstrated its efficacy in managing gastric discomfort caused by Helicobacter pylori infection [12] and mitigating the side effects induced by chemotherapeutic drugs. Helicobacter pylori-induced gastric cancer is believed to be a result of chronic inflammation that can lead to atrophic gastritis, gastric intestinal metaplasia, dysplasia, and eventually gastric adenocarcinoma [12, 13].
HZQYF is composed of Chebulae Fructus, Ficus hirta Vahl, and Cloves. Chebulae Fructus has been considered to possess antioxidant, hepatoprotective, neuroprotective, gastroprotective, antidiabetic, and anti-inflammatory activities [14]. Additionally, our previous study proved that Chebulae Fructus inhibited Helicobacter pylori-induced inflammatory response [15]. Cloves, also known as Syzygium aromaticum (L.) Merr. & L.M Perry, is traditionally used to support gastrointestinal function, alleviate pain, and treat stomach disorders such as vomiting, flatulence, and nausea [16]. Ficus hirta Vahl is the dried root of Ficus hirta Vahl. Studies have shown that Ficus hirta Vahl has the functions of liver protection, cancer prevention, and enhancement of immunological function [17,18,19]. Another study proved that it exerts antiproliferative and growth-inhibitory effects on Hela cells [20].
Herein, this study aimed to investigate its anti-gastric cancer effects and explore its possible mechanisms.
Material and Methods
The preparation of HZQYF water extracts
Chebulae Fructus and Ficus hirta Vahl (Root) were obtained from Guangzhou Zhining Pharmaceutical Co., Ltd. Cloves were offered by Guangzhou Junyuan Chenxiangshan Traditional Chinese Medicine Herbal Pieces Co., Ltd. 10 g HZQYF (60% Ficus hirta Vahl, 30% Chebulae Fructus and 10% Cloves) were extracted with tenfold water and boiled three times for 1 h at 90 ℃, then filtered, concentrated, and freeze-dried to powder. HZQYF water extracts were stored at − 20 ℃ and deposited in the International Pharmaceutical Engineering Lab of Shandong Province.
Ultra-high performance liquid chromatography-MS/MS (UHPLC-MS/MS)
As described in our previously published article [22], 1 mg/mL HZQYF water extract was filtered through 0.22 μm membrane and analyzed by UHPLC-MS/MS (Vanquish Q Exactive, Thermo Fisher, USA). The mobile phase was water-acetic acid (100:0.2, solvent A) and methanol (solvent B). The gradient condition was 0 min: 95% A, 5% B; 5 min: 85% A, 15% B; 10 min: 75% A, 25% B; 20 min: 40% A, 60% B; 35-45 min: 10% A, 90% B; 45.1- 60 min: 95% A, 5% B. The flow rate was 0.2 mL/min and the detected UV spectra was 270 nm. The Chromatography Column was ACQUITY UPLC BEH C18, 2.1 * 100 mm, 1.7 μm. Monitoring mode: Full MS ddMS2; Spray voltage: (+) 3500 V (-) 3000 V; Auxiliary gas flow rate: 7 Arb; Auxiliary gas heater temperature: 300 °C; Sheath gas flow rate: 35 Arb; Capillary temperature: 325 °C.
Network Pharmacology and TCGA data analysis
Potential drug targets were screened from Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP, OB ≥ 30% and DL ≥ 0.18, http://www.tcmspw.com/tcmsp.php), SWISS Target Prediction Database (http://www.uniprot.org/) and Traditional Chinese Medicine Integrated Database (TCMID, http://www.megabionet.org/tcmid). GC RNA-seq data were obtained from The Cancer Genome Atlas-Stomach Adenocarcinoma (TCGA-STAD). Differentially expressed genes (DEGs) of GC were analyzed by the R Foundation with a fold change cut-off value of 1.3 and a P value of less than 0.05. The Venn Diagram was drawn online (https://bioinformatics.psb.ugent.be/webtools/Venn/). The protein–protein association network was analyzed by the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database in 2023 (https://cn.stringdb.org/cgi/input?sessionId=b4apxqd43I6Z&input_page_show_search=on).
GO and KEGG analysis
Enrichment GO term (BP/CC/MF) and KEGG were plotted by https://www.bioinformatics.com.cn (last accessed on 10 Nov 2023), an online platform for data analysis and visualization [21].
Reactome pathway
The Reactome Pathway was analyzed using Analysis Tools (https://reactome.org/Â ) [22].
Cell viability
Cell viability was conducted by CCK- 8 assay [23]. GC cells (BGC- 823 cells and SGC- 7901 cells) were cultured in RMPI 1640 medium containing 10% FBS in a 37℃ CO₂ incubator with a humid atmosphere. 6000 cells of BGC- 823 and SGC- 7901 were seeded in 96-well plates overnight and co-incubated with different concentrations of HZQYF (0–800 µg/mL) for 48 h. Then 10% CCK- 8 solutions were added into the medium and incubated for 2 h at 37 ℃. The process details of the drug preparation were as follows: Initially, 10 mg of HZQYF water extract powder was precisely weighed and dissolved in 1 mL of distilled water. Subsequently, the solution was filtered through a 0.22 μm membrane to obtain a filtrate for subsequent use. Finally, the 10 mg/mL stock solution of HZQYF was serially diluted with RMPI 1640 medium containing 10% FBS to reach the desired concentrations of 50, 100, 200, 400, and 800 µg/mL. The control group was treated in a way similar to that of the stock HZQYF solution. Specifically, it was initially treated with an equal volume of distilled water and further diluted with RMPI 1640 medium containing 10% FBS.
Cell colony formation
500 cells were seeded and cultured in a 12-well plate overnight. Different concentrations of HZQYF (0, 50, 100, 200 µg/mL) were treated with the GC cells. The medium was changed every three days and the cells were cultured until the number of clones was visible and then fixed with paraformaldehyde for 15 min, stained with 0.1% crystal violet, and photographed.
Data analysis
Statistical analyses were conducted using GraphPad Prism 8 (GraphPad Software, San Diego, CA, USA). Data are expressed as mean ± standard deviation (SD). Differences between groups were assessed using an unpaired two-tailed Student’s t-test, with significance levels denoted as *P < 0.05 and **P < 0.01. To ensure robustness and reproducibility, all experiments were performed in triplicate.
Results
The main compounds were identified by UHPLC-MS/MS
In order to determine the main components of the drug, we performed a UHPLC-MS/MS analysis [22]. The results demonstrated that the primary compounds were identified and characterized, namely Shikimic acid, Chebulic acid, Quinic Acid, Punicalin, Gallic Acid, Galloyl-glucose, Galloylquinic acid, HHDP-glucose, Digalloyl glucoside, HHDP-hexoside, Vanillic acid glucose, Corilagin, Magnolioside, Brevifolin-carboxylic acid, Chebulanin, Trigalloyl-glucoside, HHDP-galloyl glucose, Galloyl chebuloyl-HHDP Glucose, Tetragalloyl glucose, Chebulinic acid, Quercetin- 3-o-Glucuronide, Ellagic acid, Kaempferol, kaempferol- 3-Oglucuronide, Quercetin- 3-Oglucuronide, 6’’-methylester, 3-Methylquercetin (Isorhamnetin), Psoralen, Angelicin and Bergapten (Fig. 1, S-Table 1).
The potential targets of HZQYF and GC were identified via network pharmacology and transcript data analysis
As shown in Fig. 2 A, 289 drug targets of HZQYF were screened, consisting of targets of Chebulae Fructus, Ficus hirta Vahl, and Cloves, respectively. Subsequently, 260 genes were co-expressed between HZQYF and GC targets (Fig. 2B). The PPI network diagram of these genes is shown in Fig. 2C.
The potential targets of HZQYF were identified through network pharmacology. A. Venn diagram of 289 targets of DX, HZ and WZMT screened from Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP, OB ≥ 30% and DL ≥ 0.18, http://www.tcmspw.com/tcmsp.php), SWISS Target Prediction Database (http://www.uniprot.org/) and Traditional Chinese Medicine Integrated Database (TCMID, http://www.megabionet.org/tcmid). B. Venn diagram of 289 targets from HZQYF and Differentially expressed genes (DEGs) of GC. GC RNA-seq data were obtained from The Cancer Genome Atlas-Stomach Adenocarcinoma (TCGA-STAD). DEGs of GC were analyzed by the R Foundation with a fold change cut-off value of 1.3 and a P value of less than 0.05. C. The PPI network of 260 co-expressed targets from HZQYF and DEGs of GC. FF represents HZQYF; DX represents Cloves; HZ represents Chebulae Fructus; WZMT represents Ficus hirta Vahl; GC represents gastric cancer; PPI represents protein-protein interaction.
GO and KEGG pathway analysis
We performed pathway prediction analyses to explore the pathways that may be enriched for these drug-disease targets. From the results of GO analysis, we found that the HZQYF anti-GC effect may be achieved through the regulation of steroid metabolic process, postsynaptic membrane potential, GABA-A/GABA receptor complex, chloride channel complex, postsynaptic membrane, steroid activity, extracellular ligand-gated ion channel activity, and transmitter-gated ion channel activity (Fig. 3A). KEGG results indicated that it may control reactive oxygen species, GABAergic synapse, Nitrogen metabolism (Fig. 3B).
The 260 DEGs were analyzed via GO and KEGG pathway. A. GO analysis. B. KEGG pathway. Enrichment GO term (BP/CC/MF) and KEGG were plotted by https://www.bioinformatics.com.cn (last accessed on 10 Nov 2023). DEGs represents differentially expressed genes; GO represents Gene Ontology; KEGG represents Kyoto Encyclopedia of Genes and Genomes
Reactome analysis
In order to further explore the relationship between HZQYF and GC, Reactome analysis was conducted. As displayed in Fig. 4, the results of enrichment analysis suggested that the anti-GC effect of HZQYF might be related to the signal transduction, translocation of olfactory receptors, neutrophil degranulation.
260 DEGs were analyzed via Reactome pathway. The Reactome Pathway was analyzed using Analysis Tools (https://reactome.org/)
The Inhibitory effect of HZQYF on gastric cancer cells
Next, to investigate its active role, we examined the anti-gastric cancer activity of HZQYF. The experimental results demonstrated that HZQYF at concentrations of 50, 100, 200, 400, and 800 µg/mL significantly inhibited the viability of GC cells (Fig. 5A and 5B). Additionally, HZQYF at concentrations of 50, 100, and 200 µg/mL suppressed the clonogenic formation ability of GC cells (Fig. 5C).
HZQYF inhibits cell viability and colony formation of gastric cancer. A. Cell viability was analyzed by CCK- 8 assay. Cells were treated with HZQYF at concentrations of 0, 50, 100, 200, 400, and 800 µg/mL for 48 h. B. Observation of changes in cell morphology after 48 h of HZQYF treatment under the microscope (100X). C. Cloning formation experiment after HZQYF treatment. HZQYF represents Hezi Qingyou Formula; CCK- 8 represents Cell Counting Kit- 8. FF50, FF100, and FF200 respectively denote cells that underwent treatment with 50 µg/mL, 100 µg/mL, and 200 µg/mL of HZQYF
Discussion
Network pharmacology and bioinformatics provide high efficiency for studying the core pathways and targets of their actions, as well as laying the theoretical foundation for subsequent precision therapy. The combined use of network pharmacology and bioinformatics offers favorable evidence in our search for the effects and targets of herbal compounds against gastric cancer. Moreover, this approach provides a better understanding of the valuable efficiency of screening based on big data.
Gastric cancer is a highly lethal malignant tumor [24]. The treatment for advanced gastric cancer has evolved significantly, particularly with the advent of immune checkpoint inhibitors combined with chemotherapy, which has shown remarkable survival benefits for patients with positive PD-L1 expression. Combining immune checkpoint inhibitors and chemotherapy can remarkably prolong survival [25,26,27]. However, the over-activation of the immune system may attack the normal tissues and organs of the body, leading to immune-related adverse reactions, such as cytokine release syndrome, which may compromise patient safety [28]. Furthermore, the efficacy of immune checkpoint inhibitors is predominantly observed in PD-L1-positive patients, while those with PD-L1-negative expression derive limited benefit [29]. Not all advanced gastric cancer patients can benefit from immunotherapy, and it is necessary to accurately screen the appropriate patient population [30]. In addition, the significant toxic side effects and substantial financial burden associated with current chemotherapeutic and immunotherapeutic agents have prompted the exploration of alternative therapeutic strategies. In light of these challenges, there is growing interest in exploring alternative or adjunctive therapies, particularly traditional herbal formulas, which may offer a safer and more cost-effective approach. It has been reported that certain Chinese herbal drugs, like Sishen Jiedu Decoction, Pingxiao Tablet, Fuzheng Guben Granule, and Buyang Huanwu Tang, are capable of enhancing the five-year survival rates of patients afflicted with upper digestive tract cancers [31].
As a clinical formulation, Hezi Qingyou Formula (HZQYF) represents a novel and promising candidate. While standard treatments like chemotherapy and immunotherapy remain cornerstone therapies, HZQYF offers a complementary approach that may address some of their limitations, such as toxicity and high costs. In this study, we present the first experimental evidence demonstrating the anti-gastric cancer activity of HZQYF in vitro, underscoring its potential as a promising therapeutic agent. To further explore its efficacy and elucidate its mechanisms of action, future studies directly comparing HZQYF with chemotherapy or immunotherapy are essential. Such comparative analyses will not only validate its therapeutic potential but also pave the way for its integration into comprehensive gastric cancer treatment regimens, offering a complementary or alternative approach to current therapies.
In this study, we initially identified potential drug targets of HZQYF by analyzing three medicinal herbs—Chebulae Fructus, Ficus hirta Vahl, and Cloves—using network pharmacology. To ensure the relevance of these targets to gastric cancer, we integrated them with differentially expressed genes identified from TCGA-STAD gastric cancer tissue sequencing data. Through this combined analysis, we identified 260 shared target genes that are likely to play critical roles in gastric cancer pathophysiology. These targets were further prioritized based on their involvement in key biological processes and signaling pathways associated with gastric cancer. This approach ensures that the selected targets are not only relevant to the pharmacological action of HZQYF but also directly linked to the molecular mechanisms underlying gastric cancer progression.
Meanwhile, based on the results of big data, it was predicted that the core probable function of HZQYF might be regulating reactive oxygen species (ROS), GABAergic synapse, and nitrogen metabolism. ROS, which includes superoxide anions, hydrogen peroxide, and hydroxyl radicals, are essential for maintaining normal physiological functions when balanced within and outside cells. However, excessive ROS production or impaired antioxidant defense systems can lead to oxidative stress, closely associated with tumor initiation and progression, including gastric cancer [32]. Excessive ROS can cause oxidative damage to DNA, proteins, and lipids, promoting abnormal cell proliferation and carcinogenesis. Additionally, ROS are involved in various signaling pathways related to gastric cancer, such as inflammation, cell apoptosis, and angiogenesis, further promoting gastric cancer development [32, 33]. Additionally, ROS plays a pivotal role in modulating the PI3 K/AKT signaling pathway. By precisely controlling ROS levels, this pathway can be inhibited, leading to the induction of apoptosis and thereby effectively curbing the advancement of cancer [34]. Elevated ROS levels can induce oxidative stress, resulting in DNA damage and genomic instability, further activating PI3 K/AKT signaling to drive tumor growth [35]. Therefore, controlling ROS levels and maintaining the oxidative-antioxidative balance are crucial for the prevention and treatment of gastric cancer.
GABAergic synapse can impact gastric cancer through its potential influence on the neurotransmitter gamma-aminobutyric acid (GABA) and its associated synaptic pathways [36]. GABA, known as an inhibitory neurotransmitter in the central nervous system, has been observed to affect non-neuronal tissues, including the gastrointestinal system. A prior study indicated that GABA and its receptors may influence various cancer-related processes, such as cell proliferation, apoptosis, migration, and invasion [37]. Moreover, the presence of GABA receptors in gastric cancer cells suggests a potential association between GABAergic signaling and the development of gastric cancer [38]. What’s more, GABA receptors could act as a novel therapeutic target in cancer [39, 40]. GABA has been shown to stimulate the proliferation of human gastric cancer cells through both autocrine and paracrine mechanisms mediated by GABAAR. This process subsequently triggers the activation of the MAPK/ERK signaling pathway and upregulates cyclin D1 expression [41]. Therefore, a comprehensive investigation of the GABA signaling pathway is of paramount importance, as HZQYF may serve as a potential therapeutic target that plays a crucial regulatory role in this pathway. Further studies will be conducted to elucidate its molecular mechanisms and explore its therapeutic potential in gastric cancer treatment.
Nitrogen metabolism can impact gastric cancer through various mechanisms [42]. For instance, changes in nitrogen metabolism can influence the synthesis of nucleotides and amino acids, which are crucial for cell growth and proliferation [43]. Additionally, nitrogen metabolism contributes to the production of reactive nitrogen species, which may lead to oxidative stress and DNA damage, potentially promoting cancer development. Moreover, nitrogen-containing compounds like nitrosamines, formed as a result of nitrogen metabolism in the human body, are implicated in the development of gastric cancer. Overall, disruptions in nitrogen metabolism can affect cellular processes and contribute to the molecular pathways involved in the initiation and progression of gastric cancer.
In addition, we predicted that its possible components that exert anti-tumor effects are likely to be chebulic acid, quinic acid, gallic acid, corilagin, tetragalloyl glucose, chebulinic acid, ellagic acid, kaempferol, isorhamnetin, psoralen, angelicin and bergapten. Studies have shown that gallic acid is a potential anticancer agent [44], inducing cell apoptosis [45], inhibiting cell migration and invasion via blocking the AKT/small GTPase signals pathway, and inhibition NF-kappaB activity [46]. Additionally, research has demonstrated that corilagin and tetragalloyl glucose can induce cancer cell apoptosis [47,48,49,50], ellagic acid can suppress the migration and invasion of GC cells [51,52,53], kaempferol can promote autophagy and cell death, and inhibit tumor growth [54,55,56,57,58]. Isorhamnetin has been found to inhibit proliferation and invasion and induce apoptosis [59,60,61,62]. Research revealed that psoralen increases chemotherapeutic sensitivity [63, 64] and angelicin inhibits liver cancer growth [65]. Additionally, research has found that bergapten has anti-cancer and anti-bacterial properties, reduces cell viability, induces cell cycle arrest, and exerts potent anticancer and apoptotic effects through autophagy [66,67,68,69,70,71].
Our study provides a rational theoretical basis for clinical prescription against tumors. Given the current research context and the scope of our study, we mainly resorted to in-silico methods. Network pharmacology, which can analyze complex drug-target-disease interactions at a systems level, helped us map potential pathways and molecular mechanisms. Transcriptomic analysis, meanwhile, revealed gene expression profiles relevant to our research, allowing us to identify key genes and regulatory networks. These in-silico techniques provided valuable clues and emphasized the predictive value of our findings as a basis for future experiments.
However, this article has some shortcomings. First, pathway-based validation should be performed. At present, pathway-based experimental validation is lacking. In the forthcoming work, we have devised a plan to carry out a comprehensive series of pathway-centered experiments. These will include assays for ROS modulation and investigations into the GABAergic synapse.
Additionally, we have only carried out preliminary investigations through cell experiments. To fully validate the therapeutic efficacy of HZQYF against GC and its potential for clinical translation, further in vivo studies are essential. In the subsequent research, we will conduct animal models. Specifically, we plan to select appropriate animal models for gastric cancer, such as the mouse xenograft tumor model. Prior to the experiments, we will accurately convert the clinical dose into the dosing doses for mice or rats based on well-established conversion principles and meticulously design three dose groups: low, medium, and high. This approach will allow us to systematically analyze the impact of different doses of HZQYF on the treatment effect, with particular emphasis on enhancing efficacy and monitoring the potential toxicity. During the experiments, we will conduct them strictly in accordance with the standard operating procedures. By observing the tumor growth curve, evaluating the changes in tumor volume and weight, and detecting relevant biomarkers and other indicators, we will comprehensively explore the inhibitory effect of HZQYF on gastric cancer in the in vivo environment and its underlying mechanisms. Simultaneously, we will also pay close attention to the overall physiological status of the animals, including changes in body weight, diet, and activity. This comprehensive assessment will help us determine the safety and tolerability of HZQYF.
In addition, advanced molecular biology techniques will be employed. Specifically, gene knockout and overexpression models will be utilized to deliberately disrupt specific pathways. By doing so, the consequent phenotypic alterations can be closely monitored and recorded, which will serve as crucial indicators of the functional changes within the biological system. Subsequently, high-throughput screening approaches will be implemented. 16S rRNA sequencing and proteomics analysis, in particular, will be carried out. These techniques will enable us to meticulously and thoroughly characterize the molecular modifications that occur within the pathways. It is not only anticipated to effectively fill the existing gap in pathway-based experimental validation, which has been a limitation in our current study, but also to generate more accurate, reliable, and conclusive outcomes. Consequently, this will significantly enhance and strengthen the scientific foundation. Moreover, it will expand the translational potential of our research, making it more likely to have practical applications and impacts in relevant fields. Meanwhile, the clinical formula is only based on the in-hospital preparation being used, and we will follow up with multi-clinical center experiments to explore the possibility of developing it into a new drug.
For the development of clinical prescriptions into new drugs for research, there is still a lot of work that needs to be done to follow up with further research. The translation of HZQYF into clinical practice presents both promising prospects and numerous considerations. Navigating the complex pharmaceutical regulatory network is an extremely challenging task. The approval process demands comprehensive documentation, ranging from detailed preclinical data to well-structured clinical trial protocols. To meet regulatory standards, strict adherence to good laboratory practice and good clinical practice is essential. For instance, during the submission of a new drug application, exhaustive information regarding HZQYF's chemical composition analysis, entire production process, and pharmacological properties must be provided. This process is time-consuming and resource-intensive and requires the relevant personnel to have a deep understanding of the constantly evolving regulatory landscape.
Transitioning from small-scale laboratory preparations to large-scale industrial production poses a series of difficulties. Ensuring the stability of raw material supply in large-scale production is crucial, as minor alterations can significantly impact the drug's final efficacy and safety. Scaling up production necessitates comprehensive and meticulous optimization of all aspects to maintain consistent product quality. Key factors include the selection of appropriate production equipment, precise control of temperature and humidity throughout production, and proper establishment of post-production storage conditions. If the formulation originated from a small laboratory, challenges such as batch-to-batch reproducibility and cost-effective procurement of high-quality raw materials must be overcome during industrialization.
A thorough investigation of potential side effects is of utmost importance. Cell and animal experiments conducted in the early stage, as well as any available human data, require careful analysis. Understanding the frequency and severity of potential side effects is not only vital for patient safety but also critical for meeting regulatory requirements and enhancing the overall acceptance of HZQYF in the clinical practice environment.
The translation of HZQYF into clinical practice necessitates a rigorous approach to dosage determination and safety evaluation, both of which are critical to ensuring therapeutic efficacy and patient safety. To systematically evaluate its efficacy, animal studies will employ the body-surface area normalization method to calculate species-specific dosages, ensuring translational relevance. Specifically, the drug’s effects will be investigated at 0.5 ×, 1 ×, and 2 × the human equivalent dose, enabling a comprehensive analysis of the dose–response relationship and providing critical insights for optimizing therapeutic dosages in future clinical trials. Safety assessment is equally paramount, and a robust monitoring framework has been designed for clinical implementation. This includes routine biochemical profiling to evaluate hepatic and renal function, as well as vigilant monitoring for potential allergic reactions. Furthermore, the safety protocol aligns with guidelines from authoritative regulatory bodies such as the FDA and EMA, ensuring compliance with international standards. By integrating dose–response characterization with rigorous safety evaluation and regulatory adherence, this approach establishes a scientifically validated and globally compliant pathway for translating HZQYF from preclinical research to clinical application, thereby bridging the gap between traditional medicine and modern therapeutic development.
Although the translation of HZQYF into clinical practice is faced with hardships and challenges, by adopting a multi-pronged comprehensive strategy to address issues in key areas such as regulation, formulation, and safety, the door to successful clinical application can be opened smoothly.
Conclusion
Based on this, we found that the clinical prescription HZQYF has anti-gastric cancer activity in vitro, while the core pathway proteins of its action may regulation of GABAergic synapse and nitrogen metabolism. Additionally, its core active ingredients are mainly chebulic acid, quinic acid, gallic acid, corilagin, tetragalloyl glucose, chebulinic acid, ellagic acid, kaempferol, isorhamnetin, psoralen, angelicin and bergapten. This provides a theoretical basis for the development of clinical formulas into new drugs.
Data availability
The datasets used and analyzed during the current study are available from the corresponding author on reasonable request.
Abbreviations
- HZQYF:
-
Hezi Qingyou Formula
- GC:
-
Gastric cancer
- GO:
-
Gene Ontology
- KEGG:
-
Kyoto Encyclopedia of Genes and Genomes
- STRING:
-
Search Tool for the Retrieval of Interacting Genes/Proteins
- TCM:
-
Traditional Chinese medicine
- TCGA-STAD:
-
The Cancer Genome Atlas-Stomach Adenocarcinoma
- TCMID:
-
Traditional Chinese Medicine Integrated Database
- TCMSP:
-
Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform
- DEGs:
-
Differentially expressed genes
- UHPLC-MS/MS:
-
Ultra-high performance liquid chromatography coupled with tandem mass spectrometer
- ROS:
-
Reactive oxygen species
- GABA:
-
Gamma-aminobutyric acid
References
Alagesan P, Goodwin JC, Garman KS, Epplein M. Cancer Progress and Priorities: Gastric Cancer. Cancer Epidemiol Biomarkers Prev. 2023;32(4):473–86.
Karim F, Amin A, Liu M, Vishnuvardhan N, Amin S, Shabbir R, Swed B, Khan U. Role of Checkpoint Inhibitors in the Management of Gastroesophageal Cancers. Cancers (Basel). 2023;15(16):4099.
Sexton RE, Al Hallak MN, Diab M, Azmi AS. Gastric cancer: a comprehensive review of current and future treatment strategies. Cancer Metastasis Rev. 2020;39(4):1179–203.
Murphy A, Kelly RJ. Immunotherapy in upper GI malignancies. Curr Treat Options Oncol. 2015;16(5):20.
Chongqing T, Liubao P, Xiaohui Z, Jianhe L, Xiaomin W, Gannong C, Siying W, Lihui O, Ziying Z. Cost-utility analysis of the newly recommended adjuvant chemotherapy for resectable gastric cancer patients in the 2011 Chinese National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology: Gastric Cancer. Pharmacoeconomics. 2014;32(3):235–43.
Lee JH, Kim KM, Cheong JH, Noh SH. Current management and future strategies of gastric cancer. Yonsei Med J. 2012;53(2):248–57.
Shah MA, Kennedy EB, Alarcon-Rozas AE, Alcindor T, Bartley AN, Malowany AB, Bhadkamkar NA, Deighton DC, Janjigian Y, Karippot A, et al. Immunotherapy and Targeted Therapy for Advanced Gastroesophageal Cancer: ASCO Guideline. J Clin Oncol. 2023;41(7):1470–91.
Liu T, Bai Y, Lin X, Li W, Wang J, Zhang X, Pan H, Bai C, Bai L, Cheng Y, et al. First-line nivolumab plus chemotherapy vs chemotherapy in patients with advanced gastric, gastroesophageal junction and esophageal adenocarcinoma: CheckMate 649 Chinese subgroup analysis. Int J Cancer. 2023;152(4):749–60.
Shih WT, Yang PR, Shen YC, Yang YH, Wu CY. Traditional Chinese Medicine Enhances Survival in Patients with Gastric Cancer after Surgery and Adjuvant Chemotherapy in Taiwan: A Nationwide Matched Cohort Study. Evid Based Complement Alternat Med. 2021;2021:7584631.
Wu CE, Xue WW, Zhuang YW, Ding DW, Zhou JY, Liu SL, Wang RP, Shu P. A clinical study on the efficacy of Yiqi Huayu Jiedu decoction for reducing the risk of postoperative recurrence and metastasis of gastric cancer: Protocol for a multicenter, randomized, double-blind, placebo-controlled trial. Medicine (Baltimore). 2020;99(48): e23417.
Pan X, Tao H, Nie M, Liu Y, Huang P, Liu S, Sun W, Wu J, Ma T, Dai A, et al. A clinical study of traditional Chinese medicine prolonging the survival of advanced gastric cancer patients by regulating the immunosuppressive cell population: A study protocol for a multicenter, randomized controlled trail. Medicine (Baltimore). 2020;99(16): e19757.
Hsiung HY, Fann JC, Yen AM, Chen SL, Chiu SY, Ku TH, Liu TY, Chen HH, Lin MW. Stage-specific Dietary Factors Associated with the Correa Multistep and Multifactorial Process of Human Gastric Carcinogenesis. Nutr Cancer. 2016;68(4):598–610.
Correa P. Human gastric carcinogenesis: a multistep and multifactorial process–first American cancer society award lecture on cancer epidemiology and prevention. Cancer Res. 1992;52(24):6735–40.
Nigam M, Mishra AP, Adhikari-Devkota A, Dirar AI, Hassan MM, Adhikari A, Belwal T, Devkota HP: Fruits of Terminalia chebula Retz.: A review on traditional uses, bioactive chemical constituents and pharmacological activities. Phytother Res 2020, 34(10):2518–2533.
Ou L, Liu HR, Shi XY, Peng C, Zou YJ, Jia JW, Li H, Zhu ZX, Wang YH, Su BM et al: Terminalia chebula Retz. aqueous extract inhibits the Helicobacter pylori-induced inflammatory response by regulating the inflammasome signaling and ER-stress pathway. J Ethnopharmacol 2024, 320:117428.
Batiha GE, Alkazmi LM, Wasef LG, Beshbishy AM, Nadwa EH, Rashwan EK: Syzygium aromaticum L. (Myrtaceae): Traditional Uses, Bioactive Chemical Constituents, Pharmacological and Toxicological Activities. Biomolecules 2020, 10(2):202.
Au DT, Chen H, Jiang Z, Zhao Z. A novel method to identify the Chinese herbal medicine Wuzhimaotao by quantification of laticifers. Microsc Res Tech. 2009;72(4):293–8.
Cai QY, Chen HB, Cai SQ, Zhao ZZ, Ruan M, Jia FL, Ou T, Zhang BX. Effect of roots of Ficus hirta on cocaine-induced hepatotoxicity and active components. Zhongguo Zhong Yao Za Zhi. 2007;32(12):1190–3.
Zhou FY, Quan T, Xiang SJ, Li H, Chen HY, Yang YX, Li SS. Metabolomic Mechanisms of Radix Fici Hirtae against Carbon Tetrachloride-Induced Acute Liver Damage in Mice. Evid Based Complement Alternat Med. 2022;2022:9157465.
Zeng YW, Liu XZ, Lv ZC, Peng YH: Effects of Ficus hirta Vahl. (Wuzhimaotao) extracts on growth inhibition of HeLa cells. Exp Toxicol Pathol 2012, 64(7–8):743–749.
Tang D, Chen M, Huang X, Zhang G, Zeng L, Zhang G, Wu S, Wang Y. SRplot: A free online platform for data visualization and graphing. PLoS ONE. 2023;18(11): e0294236.
Gillespie M, Jassal B, Stephan R, Milacic M, Rothfels K, Senff-Ribeiro A, Griss J, Sevilla C, Matthews L, Gong C, et al. The reactome pathway knowledgebase 2022. Nucleic Acids Res. 2022;50(D1):D687–92.
Cai L, Qin X, Xu Z, Song Y, Jiang H, Wu Y, Ruan H, Chen J. Comparison of Cytotoxicity Evaluation of Anticancer Drugs between Real-Time Cell Analysis and CCK-8 Method. ACS Omega. 2019;4(7):12036–42.
Petryszyn P, Chapelle N, Matysiak-Budnik T. Gastric Cancer: Where Are We Heading? Dig Dis. 2020;38(4):280–5.
Chenard-Poirier M, Smyth EC. Immune Checkpoint Inhibitors in the Treatment of Gastroesophageal Cancer. Drugs. 2019;79(1):1–10.
Zhang Z, Xie T, Zhang X, Qi C, Shen L, Peng Z. Immune checkpoint inhibitors for treatment of advanced gastric or gastroesophageal junction cancer: Current evidence and future perspectives. Chin J Cancer Res. 2020;32(3):287–302.
Yao G, Yuan J, Duan Q, Tan Y, Zhang Q, Chen D, Chen J. Immunoneoadjuvant therapy with immune checkpoint inhibitors of gastric cancer: an emerging exemplification : Immunoneoadjuvant therapy of gastric cancer. Invest New Drugs. 2024;42(1):1–13.
Zhang X, You Y, Zhang P, Wang Y, Shen F. Cytokine release syndrome caused by immune checkpoint inhibitors: a case report and literature review. Future Sci OA. 2024;10(1):2422786.
Chen C, Zhang F, Zhou N, Gu YM, Zhang YT, He YD, Wang L, Yang LX, Zhao Y, Li YM. Efficacy and safety of immune checkpoint inhibitors in advanced gastric or gastroesophageal junction cancer: a systematic review and meta-analysis. Oncoimmunology. 2019;8(5): e1581547.
Kono K, Nakajima S, Mimura K. Current status of immune checkpoint inhibitors for gastric cancer. Gastric Cancer. 2020;23(4):565–78.
Huang AS, Wu J, Amin A, Fu XQ, Yu ZL. Traditional Chinese medicine in treating upper digestive tract cancers. Mol Cancer. 2024;23(1):250.
Biagioni A, Peri S, Versienti G, Fiorillo C, Becatti M, Magnelli L, Papucci L. Gastric Cancer Vascularization and the Contribution of Reactive Oxygen Species. Biomolecules. 2023;13(6):886.
Wu S, Chen Y, Chen Z, Wei F, Zhou Q, Li P, Gu Q. Reactive oxygen species and gastric carcinogenesis: The complex interaction between Helicobacter pylori and host. Helicobacter. 2023;28(6): e13024.
Cao J, Xu D, Wang D, Wu R, Zhang L, Zhu H, He Q, Yang B. ROS-driven Akt dephosphorylation at Ser-473 is involved in 4-HPR-mediated apoptosis in NB4 cells. Free Radic Biol Med. 2009;47(5):536–47.
Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev Drug Discov. 2009;8(7):579–91.
Huang D, Alexander PB, Li QJ, Wang XF. GABAergic signaling beyond synapses: an emerging target for cancer therapy. Trends Cell Biol. 2023;33(5):403–12.
Young SZ, Bordey A. GABA’s control of stem and cancer cell proliferation in adult neural and peripheral niches. Physiology (Bethesda). 2009;24:171–85.
Matuszek M, Jesipowicz M, Kleinrok Z. GABA content and GAD activity in gastric cancer. Med Sci Monit. 2001;7(3):377–81.
Bhattacharya D, Gawali VS, Kallay L, Toukam DK, Koehler A, Stambrook P, Krummel DP, Sengupta S. Therapeutically leveraging GABA(A) receptors in cancer. Exp Biol Med (Maywood). 2021;246(19):2128–35.
Sawaki K, Kanda M, Baba H, Inokawa Y, Hattori N, Hayashi M, Tanaka C, Kodera Y. Gamma-aminobutyric Acid Type A Receptor Subunit Delta as a Potential Therapeutic Target in Gastric Cancer. Ann Surg Oncol. 2023;30(1):628–36.
Maemura K, Shiraishi N, Sakagami K, Kawakami K, Inoue T, Murano M, Watanabe M, Otsuki Y. Proliferative effects of gamma-aminobutyric acid on the gastric cancer cell line are associated with extracellular signal-regulated kinase 1/2 activation. J Gastroenterol Hepatol. 2009;24(4):688–96.
Kurmi K, Haigis MC. Nitrogen Metabolism in Cancer and Immunity. Trends Cell Biol. 2020;30(5):408–24.
Shen J, Wang RT, Wang LW, Xu YC, Wang XR. A novel genetic polymorphism of inducible nitric oxide synthase is associated with an increased risk of gastric cancer. World J Gastroenterol. 2004;10(22):3278–83.
Jiang Y, Pei J, Zheng Y, Miao YJ, Duan BZ, Huang LF. Gallic Acid: A Potential Anti-Cancer Agent. Chin J Integr Med. 2022;28(7):661–71.
Tsai CL, Chiu YM, Ho TY, Hsieh CT, Shieh DC, Lee YJ, Tsay GJ, Wu YY. Gallic Acid Induces Apoptosis in Human Gastric Adenocarcinoma Cells. Anticancer Res. 2018;38(4):2057–67.
Ho HH, Chang CS, Ho WC, Liao SY, Lin WL, Wang CJ. Gallic acid inhibits gastric cancer cells metastasis and invasive growth via increased expression of RhoB, downregulation of AKT/small GTPase signals and inhibition of NF-kappaB activity. Toxicol Appl Pharmacol. 2013;266(1):76–85.
Yang MH, Baek SH, Hwang ST, Um JY, Ahn KS. Corilagin exhibits differential anticancer effects through the modulation of STAT3/5 and MAPKs in human gastric cancer cells. Phytother Res. 2022;36(6):2449–62.
Xu J, Zhang G, Tong Y, Yuan J, Li Y, Song G. Corilagin induces apoptosis, autophagy and ROS generation in gastric cancer cells in vitro. Int J Mol Med. 2019;43(2):967–79.
Zhang L, Jia B, Velu P, Wu H. Corilagin induces apoptosis and inhibits HMBG1/PI3K/AKT signaling pathways in a rat model of gastric carcinogenesis induced by methylnitronitrosoguanidine. Environ Toxicol. 2022;37(5):1222–30.
Saeki K, Hayakawa S, Noro T, Miyase T, Nakamura Y, Tanji K, Kumazawa S, Nakayama T, Isemura M. Apoptosis-inducing activity of galloyl monosaccharides in human histiocytic lymphoma U937 cells. Planta Med. 2000;66(2):124–6.
Liu Z, Huang H, Yu Y, Li L, Shi X, Wang F. Exploring the mechanism of ellagic acid against gastric cancer based on bioinformatics analysis and network pharmacology. J Cell Mol Med. 2023;27(23):3878–96.
Lu G, Wang X, Cheng M, Wang S, Ma K. The multifaceted mechanisms of ellagic acid in the treatment of tumors: State-of-the-art. Biomed Pharmacother. 2023;165: 115132.
Lim SC, Hwang H, Han SI. Ellagic Acid Inhibits Extracellular Acidity-Induced Invasiveness and Expression of COX1, COX2, Snail, Twist 1, and c-myc in Gastric Carcinoma Cells. Nutrients. 2019;11(12):3023.
Yang L, Li H, Yang M, Zhang W, Li M, Xu Y, Li J, Kang J, Zhang J, Guo S. Exploration in the Mechanism of Kaempferol for the Treatment of Gastric Cancer Based on Network Pharmacology. Biomed Res Int. 2020;2020:5891016.
Kim TW, Lee SY, Kim M, Cheon C, Ko SG. Kaempferol induces autophagic cell death via IRE1-JNK-CHOP pathway and inhibition of G9a in gastric cancer cells. Cell Death Dis. 2018;9(9):875.
Liao W, Chen L, Ma X, Jiao R, Li X, Wang Y. Protective effects of kaempferol against reactive oxygen species-induced hemolysis and its antiproliferative activity on human cancer cells. Eur J Med Chem. 2016;114:24–32.
Lin F, Luo X, Tsun A, Li Z, Li D, Li B. Kaempferol enhances the suppressive function of Treg cells by inhibiting FOXP3 phosphorylation. Int Immunopharmacol. 2015;28(2):859–65.
Song H, Bao J, Wei Y, Chen Y, Mao X, Li J, Yang Z, Xue Y. Kaempferol inhibits gastric cancer tumor growth: An in vitro and in vivo study. Oncol Rep. 2015;33(2):868–74.
Li C, Li J, Li Y, Li L, Luo Y, Li J, Zhang Y, Wang Y, Liu X, Zhou X, et al. Isorhamnetin Promotes MKN-45 Gastric Cancer Cell Apoptosis by Inhibiting PI3K-Mediated Adaptive Autophagy in a Hypoxic Environment. J Agric Food Chem. 2021;69(29):8130–43.
Li Y, Fan B, Pu N, Ran X, Lian T, Cai Y, Xing W, Sun K. Isorhamnetin Suppresses Human Gastric Cancer Cell Proliferation through Mitochondria-Dependent Apoptosis. Molecules. 2022;27(16):5191.
Ramachandran L, Manu KA, Shanmugam MK, Li F, Siveen KS, Vali S, Kapoor S, Abbasi T, Surana R, Smoot DT, et al. Isorhamnetin inhibits proliferation and invasion and induces apoptosis through the modulation of peroxisome proliferator-activated receptor gamma activation pathway in gastric cancer. J Biol Chem. 2012;287(45):38028–40.
Duan R, Liang X, Chai B, Zhou Y, Du H, Suo Y, Chen Z, Li Q, Huang X. Isorhamnetin Induces Melanoma Cell Apoptosis via the PI3K/Akt and NF-kappaB Pathways. Biomed Res Int. 2020;2020:1057943.
Choi EK, Kim HD, Park EJ, Song SY, Phan TT, Nam M, Kim M, Kim DU, Hoe KL. 8-Methoxypsoralen Induces Apoptosis by Upregulating p53 and Inhibits Metastasis by Downregulating MMP-2 and MMP-9 in Human Gastric Cancer Cells. Biomol Ther (Seoul). 2023;31(2):219–26.
Jin L, Ma XM, Wang TT, Yang Y, Zhang N, Zeng N, Bai ZG, Yin J, Zhang J, Ding GQ, et al. Psoralen Suppresses Cisplatin-Mediated Resistance and Induces Apoptosis of Gastric Adenocarcinoma by Disruption of the miR196a-HOXB7-HER2 Axis. Cancer Manag Res. 2020;12:2803–27.
Wang F, Li J, Li R, Pan G, Bai M, Huang Q. Angelicin inhibits liver cancer growth in vitro and in vivo. Mol Med Rep. 2017;16(4):5441–9.
Aslam H, Khan AU, Qazi NG, Ali F, Hassan SSU, Bungau S. Pharmacological basis of bergapten in gastrointestinal diseases focusing on H(+)/K(+) ATPase and voltage-gated calcium channel inhibition: A toxicological evaluation on vital organs. Front Pharmacol. 2022;13:1005154.
Quetglas-Llabres MM, Quispe C, Herrera-Bravo J, Catarino MD, Pereira OR, Cardoso SM, Dua K, Chellappan DK, Pabreja K, Satija S, et al. Pharmacological Properties of Bergapten: Mechanistic and Therapeutic Aspects. Oxid Med Cell Longev. 2022;2022:8615242.
Liang Y, Xie L, Liu K, Cao Y, Dai X, Wang X, Lu J, Zhang X, Li X. Bergapten: A review of its pharmacology, pharmacokinetics, and toxicity. Phytother Res. 2021;35(11):6131–47.
Lin CP, Lin CS, Lin HH, Li KT, Kao SH, Tsao SM. Bergapten induces G1 arrest and pro-apoptotic cascade in colorectal cancer cells associating with p53/p21/PTEN axis. Environ Toxicol. 2019;34(3):303–11.
Pattanayak SP, Bose P, Sunita P, Siddique MUM, Lapenna A. Bergapten inhibits liver carcinogenesis by modulating LXR/PI3K/Akt and IDOL/LDLR pathways. Biomed Pharmacother. 2018;108:297–308.
De Amicis F, Aquila S, Morelli C, Guido C, Santoro M, Perrotta I, Mauro L, Giordano F, Nigro A, Ando S, et al. Bergapten drives autophagy through the up-regulation of PTEN expression in breast cancer cells. Mol Cancer. 2015;14:130.
Acknowledgements
We thank Shanghai New Core Biotechnology Co., Ltd. (https://www.bioinformatics.com.cn, last accessed on 10 Nov 2023) for providing data analysis and visualization support.
Funding
This work was supported by the National Natural Science Foundation of China (number 81973552) and the Natural Science Foundation of Guangdong Province (number 2022 A1515012056).
Author information
Authors and Affiliations
Contributions
1—Ling Ou, Guimin Zhang, and Meicun Yao conceived and designed the experiments; 2—Zhong Feng, Ling Ou, and Hui Li performed the experiments; 3—Zhong Feng, Ling Ou, Hui Li and Yajie Hao analyzed and interpreted the data; 4—Zhong Feng, Ling Ou, Ruixia Wei, Guimin Zhang, and Meicun Yao contributed reagents, materials, analysis tools or data; 5—Zhong Feng, Ling Ou, Hui Li, Yajie Hao, Guimin Zhang, Meicun Yao wrote the paper.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Competing interest
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Feng, Z., Ou, L., Li, H. et al. Unveiling the therapeutic potential of HZQYF: exploring the inhibitory impact of a clinical herbal formula on gastric cancer through network pharmacology and transcript analysis. BMC Complement Med Ther 25, 142 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12906-025-04871-5
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12906-025-04871-5