BPG is committed to discovery and dissemination of knowledge
Randomized Clinical Trial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Surg. Dec 27, 2025; 17(12): 113364
Published online Dec 27, 2025. doi: 10.4240/wjgs.v17.i12.113364
Modified Pingwei Baohe decoction improves postoperative recovery in gastric cancer patients
Hong-Yu Ma, Yuan-Wu Chen, Department of Traditional Chinese Medicine Rehabilitation, Hebei General Hospital, Shijiazhuang 050011, Hebei Province, China
Xin Liu, Graduate School, Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
Xin Liu, Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang 050011, Hebei Province, China
Ming-Ming Zhang, Clinical Medicine Research Center, Hebei General Hospital, Shijiazhuang 050011, Hebei Province, China
Bi-Bo Tan, Third Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
ORCID number: Hong-Yu Ma (0009-0005-7602-6180); Ming-Ming Zhang (0009-0005-0795-547X); Bi-Bo Tan (0000-0001-6852-6947).
Author contributions: Conceptualization and study design were performed by Ma HY; patient recruitment, clinical implementation, and data collection were carried out by Ma HY, Chen YW, and Tan BB; laboratory testing and data analysis were conducted by Liu X and Zhang MM; the original draft of the manuscript was written by Ma HY and critically revised by Chen YW and Tan BB; all authors reviewed and approved the final version of the manuscript.
Supported by Research Program of the Hebei Provincial Administration of Traditional Chinese Medicine, No. 2025271.
Institutional review board statement: This study was conducted in accordance with the Declaration of Helsinki and was reviewed and approved by the Medical Ethics Committee of Hebei Provincial People’s Hospital and The Fourth Hospital of Hebei Medical University. All procedures involving human participants were performed only after ethical approval was obtained.
Clinical trial registration statement: This study is an exploratory clinical investigation and was not prospectively registered on a public clinical trial platform.
Informed consent statement: Written informed consent was obtained from all individual participants and their family members included in the study. Patients were fully informed about the study’s purpose, procedures, potential risks, and benefits, and they participated voluntarily.
Conflict-of-interest statement: The authors declare that they have no competing interests or financial relationships that could inappropriately influence or bias the work reported in this manuscript.
CONSORT 2010 statement: The authors have read the CONSORT 2010 Statement, and the manuscript was prepared and revised according to the CONSORT 2010 Statement.
Data sharing statement: The datasets generated and analyzed during the current study are available from the corresponding author on reasonable request, in accordance with ethical standards and participant confidentiality protections.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Hong-Yu Ma, MD, Department of Traditional Chinese Medicine Rehabilitation, Hebei General Hospital, No. 348 Heping West Road, Shijiazhuang 050011, Hebei Province, China. 505992294@qq.com
Received: August 26, 2025
Revised: September 24, 2025
Accepted: October 28, 2025
Published online: December 27, 2025
Processing time: 120 Days and 17.4 Hours

Abstract
BACKGROUND

Gastric cancer is a major global health issue, especially in East Asia. Radical gastrectomy, the primary treatment, often leads to complications like malnutrition and infection, impairing recovery. Traditional Chinese medicine may offer benefits in regulating gastrointestinal function and immunity. This study investigated the effects of a modified Pingwei Baohe decoction (MPBD) on postoperative immune function and nutritional status in gastric cancer patients.

AIM

To explore the influence of MPBD on the postoperative immune function and nutritional status of patients with gastric cancer.

METHODS

A total of 55 gastric cancer patients who underwent radical proximal gastrectomy in Hebei Provincial People's Hospital and The Fourth Hospital of Hebei Medical University from June 2023 to June 2025 were selected as the research objects and divided into the observation group (28 cases, receiving chemotherapy plus MPBD) and the control group (27 cases, receiving chemotherapy alone) at a ratio of 1:1 according to the random number table method. Inflammatory indicators, immune function, nutritional status, and gastrointestinal function recovery time were compared before and 28 days after intervention.

RESULTS

After 28 days, the observation group showed significantly lower levels of inflammatory markers (IL-6, IL-4, IL-2, IL-17A, IFN-γ, TNF-α, IL-10), neutrophil-to-lymphocyte ratio (NLR), and proportion of CONUT score > 2, alongside significantly higher levels of CD3+, CD4+, CD4+/CD8+ ratio, absolute lymphocyte count (ALC), lymphocyte percentage (LYM%), and TNF-α/IL-4 ratio compared to the control group (P < 0.05). In both groups, post-intervention levels of the aforementioned inflammatory markers, NLR, and proportion of CONUT score > 2 were significantly lower than baseline, while the immune parameters (CD3+, CD4+, CD4+/CD8+ ratio, ALC, LYM%, TNF-α/IL-4 ratio) were significantly higher (P < 0.05). Additionally, gastrointestinal function recovery times (first flatus, bowel sounds recovery, abdominal distension relief) in the observation group were significantly shorter than in the control group (P < 0.05).

CONCLUSION

MPBD significantly enhances postoperative immune function, mitigates inflammatory responses, improves nutritional status, and accelerates gastrointestinal function recovery in gastric cancer patients.

Key Words: Pingwei Baohe decoction; modified; Gastric cancer; Immune function; Nutritional status; Inflammatory response; Gastrointestinal function; Integrated traditional Chinese and Western medicine therapy

Core Tip: This randomized clinical trial demonstrates that adjunctive modified Pingwei Baohe decoction (MPBD), a traditional Chinese medicine formulation, significantly improves postoperative outcomes in gastric cancer patients. Compared to chemotherapy alone, MPBD combined with chemotherapy enhanced immune function (increased CD3+, CD4+, CD4+/CD8+ ratio), reduced inflammatory cytokines (IL-6, TNF-α), improved nutritional status (lower CONUT score), and accelerated recovery of gastrointestinal function (shorter time to first flatus and bowel sounds). MPBD represents a promising integrative therapy to support recovery after radical gastrectomy.



INTRODUCTION

Gastric cancer is a common malignant tumor of the digestive tract, with its incidence and mortality rates ranking among the highest globally, particularly in East Asia. According to 2020 statistics from the International Agency for Research on Cancer, gastric cancer accounts for 5.6% of global cancer incidence and up to 7.7% of cancer-related deaths[1], posing a severe threat to patients' lives and health. Radical gastrectomy is currently the primary treatment for gastric cancer; however, stomach removal reduces the body's storage capacity and disrupts the digestive tract structure[2]. A meta-analysis revealed that up to 68.8% of post-gastrectomy patients experience malnutrition[3], and analysis of surgical patient data showed that the incidence of postoperative incision-related infectious complications can reach 30%[4]. This indicates that surgical trauma and stress responses suppress immune function, increasing infection risks and resulting in a five-year survival rate of only 38%[5,6]. Therefore, effectively improving postoperative immune and nutritional status has become a critical focus of clinical research. Current Western medicine primarily employs nutritional supplements and immunomodulators to enhance postoperative recovery, but the adherence rate to nutritional supplements among post-gastrectomy patients is only 26.2%, with suboptimal therapeutic outcomes[7]. Traditional Chinese medicine (TCM) offers unique advantages in regulating gastrointestinal function and boosting immunity. TCM theory posits that post-gastrectomy patients exhibit a syndrome of "external excess and internal deficiency"[8]. Multiple studies have confirmed that TCM significantly reduces the risk of postoperative recurrence and metastasis in gastric cancer patients while markedly improving gastrointestinal dysfunction, shortening the time to resume normal diet from 11.84-8.88 days[9,10]. This demonstrates that TCM can effectively promote early recovery of gastrointestinal function in postoperative gastric cancer patients. Modified Pingwei Baohe decoction (MPBD) is composed of ingredients such as Atractylodes macrocephala (A. macrocephala), Magnolia officinalis (M. officinalis), dried tangerine peel, and Pinellia ternate (P. ternata). It has the effects of strengthening the spleen and stomach, aiding digestion, and clearing heat and drying dampness[11]. However, there is a lack of clinical research supporting the impact of modified MPBD on postoperative immune and nutritional status in gastric cancer patients. Thus, this study aims to explore the effects of MPBD on postoperative immune and nutritional status in gastric cancer patients, providing a new perspective for clinical intervention.

MATERIALS AND METHODS
General information

Fifty-five patients with gastric cancer who underwent radical proximal gastrectomy and were admitted to Hebei Provincial People's Hospital and The Fourth Hospital of Hebei Medical University from June 2023 to June 2025 were selected as the research subjects. The enrolled patients were numbered sequentially (01-55) and randomly divided into an observation group (n = 28) and a control group (n = 27) at a 1:1 ratio using computer-generated random numbers. The observation group consisted of 16 males and 12 females, aged 42-68 years (51.32 ± 6.11 years), with a body mass index (BMI) of 21-25 kg/m2 (22.43 ± 1.02 kg/m2), disease duration of 3-20 months (6.82 ± 3.14 months), and TCM syndrome differentiation including 11 cases of external pathogen invasion, 10 cases of food stagnation, and 7 cases of liver qi attacking the spleen. The control group comprised 14 males and 13 females, aged 44-68 years (51.61 ± 6.18 years), with a BMI of 21-24 kg/m2 (22.20 ± 1.05 kg/m2), disease duration of 4-20 months (6.74 ± 3.11 months), and TCM syndrome differentiation including 12 cases of external pathogen invasion, 9 cases of food stagnation, and 6 cases of liver qi attacking the spleen. No statistically significant differences were observed in baseline characteristics between the two groups (P > 0.05), indicating comparability.

The diagnostic criteria for gastric cancer[12] include: (1) Endoscopic confirmation of space-occupying lesions; and (2) Histological classification consistent with the World Health Organization classification of gastric cancer. In addition to meeting all the above conditions, at least one of the following criteria must be satisfied: (1) Endoscopic visualization of malignant ulcers, masses, or mucosal abnormalities; (2) Computed tomography/magnetic resonance imaging showing irregular gastric wall thickening > 1 cm with abnormal enhancement; and (3) Endoscopic ultrasound confirmation of tumor invasion beyond the muscularis mucosa (T1b or higher).

Inclusion criteria: (1) Diagnosis of early gastric cancer confirmed by gastroscopy and biopsy[12], meeting stage I-II criteria according to the UICC TNM staging system[13]; (2) Undergoing radical proximal gastrectomy; (3) Expected survival time ≥ 6 months; (4) Normal organ function confirmed by clinical evaluation; (5) No preoperative neoadjuvant chemotherapy or radiotherapy; and (6) Signed informed consent obtained from patients and their families. This study has been reviewed and approved by our hospital's medical ethics committee.

Exclusion criteria: (1) Presence of distant metastasis or locally advanced unresectable cases; (2) Patients with other concurrent malignancies; (3) BMI < 16 kg/m2 or serum albumin (ALB) < 25 g/L; (4) History of allergic reactions to any component of the investigational medication; (5) Women who are pregnant or breastfeeding; and (6) Patients with poor compliance.

Treatment methods

Patients in the control group received standard intravenous chemotherapy: On the first day after gastric cancer surgery, they were administered oxaliplatin (Jiangsu Hengrui Medicine Co., LTD., National Drug Approval Number: H20213313, Specification: 20 mL: 100 mg) via intravenous infusion at a dose of 130 mg/m2, along with oral capecitabine (Qilu Pharmaceutical Co., LTD., National Drug Approval No. H20133361, Specification: 500 mg/tablet) twice daily at 1500 mg per dose. This 7-day regimen constituted one complete treatment cycle, with a total of 4 cycles administered.

The observation group, based on the control group, took MPBD as the basic formula and modified it according to the specific symptoms of gastric cancer patients. The components of the basic formula include: 10 g of A. macrocephala, 10 g of M. officinalis, 10 g of dried tangerine peel, 10 g of P. ternata, 10 g of Poria cocos (P. cocos), 10 g of Forsythia, 10 g of wood fragrance, 10 g of roasted malt, 10 g of charred hawthorn, 10 g of roasted Six Shenqu, and 6 g of raw licorice. Adjust the prescription according to the patient's clinical symptoms: If the patient has severe chest pain and the pain spreads to both sides of the costal region, neem seeds and Corydalis yanhusuo can be added. If acid reflux symptoms occur, cuttlefish bones and Zhejiang Fritillaria can be added. If the patient has symptoms such as belching, nausea and vomiting, Perilla stems, Cyperi rhizoma, stir-fried white peony root and bamboo mushrooms can be added; If you feel chest tightness, you can add Allium victorialis, cinnamon twig and Trichosanthes kirilowii. If hematochezia occurs, the leaves of Platycladus orientalis (P. orientalis) and P. orientalis can be added. If the patient suffers from constipation, Huangshi and Zhishi can be enlarged. If the patient has a sallow complexion and feels weak, Codonopsis pilosula and Astragalus membranaceus can be added. Take one dose daily. Boil it in water to extract 300 mL of juice. Take it warm in two divided doses, 30 minutes before breakfast and dinner. Start from the first day of chemotherapy and take it continuously for 28 days.

Observation indicators and evaluation criteria

Laboratory indicator testing: Peripheral venous blood samples 2-3 mL were collected from all patients after overnight fasting using EDTA anticoagulant vacuum tubes before intervention and after 28 days of treatment. Samples were centrifuged at 3000 rpm for 5 minutes, and the supernatant was immediately transferred to -80 °C ultra-low temperature freezers for long-term storage. Serum levels of cytokines (IL-6, IL-4, IL-2, IL-17A, IFN-γ, TNF-α, and IL-10) were measured using BDTM CBA Human Th1/Th2/Th17 cytokine kits according to the manufacturer's protocol, with subsequent calculation of TNF-α/IL-4, TNF-α/IL-10, and IFN-γ/IL-4 ratios. Biochemical parameters were analyzed using an automated biochemistry analyzer: ALB was determined by bromocresol green method, total cholesterol (TC) by biuret method, while prealbumin (PA) was measured by immunoturbidimetric assay.

Immune function: Immunological assessment: Before intervention and after 28 days of treatment, 2 mL fasting peripheral venous blood samples were collected from all patients using EDTA anticoagulant vacuum tubes. T cell subsets (CD3+, CD4+, and CD8+) were analyzed by flow cytometry, with subsequent calculation of CD4+/CD8+ ratio. Detection of peripheral blood absolute lymphocyte count (ALC), lymphocyte percentage (LYM%), and neutrophil levels using a fully automated blood cell analyzer, with calculation of neutrophil-to-lymphocyte ratio (NLR) = neutrophils/ALC.

Nutritional status: Nutritional status was comprehensively evaluated using the CONUT scoring system[14] before intervention and after 28 days of treatment, calculated based on ALB, TC, and ALC parameters. The specific scoring criteria were: ALB concentration ≥ 35.0 g/L scored 0 points, 30.0-34.9 g/L scored 2 points, 25.0-29.9 g/L scored 4 points, and < 25.0 g/L scored 6 points; TC concentration ≥ 4.68 mmol/L scored 0 points, 3.64-4.67 mmol/L scored 1 point, 2.60-3.63 mmol/L scored 2 points, and < 2.60 mmol/L scored 3 points; ALC value ≥ 1.60 × 109/L scored 0 points, 1.20 × 10/L to 1.59 × 109/L scored 1 point, 0.80 × 10/L to 1.19 × 109/L scored 2 points, and < 0.80 × 109/L scored 3 points. The cumulative score from these three assessment dimensions ranged from 0-12 points. Based on the score ranges, patients' nutritional status was classified as: 0-1 indicating good nutrition, 2-4 suggesting mild malnutrition risk, 5-8 indicating moderate nutritional deficiency risk, and 9-12 representing severe nutritional deficiency.

Gastrointestinal function recovery: Record the time of the first bowel movement, defecation, and bowel sounds recovery for both groups of patients after 28 days of intervention.

Statistical analysis

The data analysis was conducted using SPSS 27.0 software. For continuous variables, mean ± SD is used for representation, independent sample t-test is used for inter group comparison, rate (%) is used for categorical variables, and χ2 test is used for comparison. The difference is statistically significant with P < 0.05.

RESULTS
Comparison of laboratory indicators between two groups

Comparison between groups: Before the intervention, there were no statistically significant differences (P > 0.05) in the serum cytokine levels (IL-6, IL-4, IL-2, IL-17A, IFN-γ, TNF-α, IL-10) between the observation group and the control group, indicating balanced baseline characteristics. After 28 days of intervention, the levels of all the aforementioned cytokines in the observation group were significantly lower than those in the control group (P < 0.05, Table 1). This indicates that MPBD combined with chemotherapy is superior to chemotherapy alone in suppressing the systemic inflammatory response.

Table 1 Between-group comparisons of laboratory parameters (mean ± SD, ng/L).
Group
Observation group (n = 28)
Control group (n = 27)
t value
P value
IL-6Before intervention35.18 ± 9.6735.72 ± 9.430.1000.835
After intervention9.81 ± 2.19a32.35 ± 9.91a11.7450.000
IL-4Before intervention6.58 ± 0.316.63 ± 0.080.8120.420
After intervention0.74 ± 0.06a4.02 ± 0.74a23.3830.000
IL-2Before intervention4.25 ± 0.824.53 ± 0.671.3840.172
After intervention0.55 ± 0.13a1.96 ± 0.21a30.0600.000
IL-17ABefore intervention10.29 ± 0.5610.34 ± 0.230.4300.669
After intervention3.25 ± 1.03a8.15 ± 0.84a19.2940.000
IFN-γBefore intervention5.36 ± 0.495.51 ± 0.730.8980.373
After intervention0.69 ± 0.17a2.58 ± 0.28a30.3850.000
TNF-αBefore intervention3.07 ± 0.513.24 ± 0.181.6360.108
After intervention1.71 ± 0.42a2.73 ± 0.33a9.9900.000
IL-10Before intervention6.37 ± 0.256.51 ± 0.790.8930.376
After intervention1.31 ± 0.32a3.59 ± 0.91a12.4580.000

Intra-group comparison: After 28 days of intervention, all cytokine levels within the observation group showed a significant decrease compared to pre-intervention levels (P < 0.001). Within the control group, while most cytokine levels also decreased significantly compared to pre-intervention levels (P < 0.05, Table 2), the IL-6 level showed no significant change (P = 0.206). This indicates that both intervention measures can modulate the inflammatory response, but the anti-inflammatory effect of the observation group is more comprehensive and consistent.

Table 2 Within-group comparisons of cytokine levels (mean ± SD, ng/L).
Group
Cytokine
Before intervention
After intervention
t value
P value
Observation group (n = 28)IL-635.18 ± 9.679.81 ± 2.1913.5400.000
IL-46.58 ± 0.310.74 ± 0.0697.8690.000
IL-24.25 ± 0.820.55 ± 0.1323.5820.000
IL-17A10.29 ± 0.563.25 ± 1.0331.7750.000
IFN-γ5.36 ± 0.490.69 ± 0.1747.6450.000
TNF-α3.07 ± 0.511.71 ± 0.4210.8930.000
IL-106.37 ± 0.251.31 ± 0.3265.9340.000
Control group (n = 27)IL-635.72 ± 9.4332.35 ± 9.911.2800.206
IL-46.63 ± 0.084.02 ± 0.7418.2210.000
IL-24.53 ± 0.671.96 ± 0.2119.0190.000
IL-17A10.34 ± 0.238.15 ± 0.8413.0660.000
IFN-γ5.51 ± 0.732.58 ± 0.2819.4730.000
TNF-α3.24 ± 0.182.73 ± 0.337.0500.000
IL-106.51 ± 0.793.59 ± 0.9112.5910.000
Comparison of immune function between two groups of patients

Comparison between groups: After the intervention, the observation group showed significantly higher levels of CD3+ and CD4+ T cells, CD4+/CD8+ ratio, ALC, LYM%, and TNF-α/IL-4 ratio compared to the control group, while the NLR was significantly lower (P < 0.05, Figure 1A-G). In addition, the TNF-α/IL-10 and IFN-γ/IL-4 ratios in the observation group were also significantly higher than those in the control group (P < 0.05, Figure 1H and I).

Figure 1
Figure 1 Comparison of immune function. A: CD3+; B: CD4+; C: CD4+/CD8+; D: Absolute lymphocyte count; E: Lymphocyte percentage; F: Neutrophil-to-lymphocyte ratio; G: TNF-α/IL-4; H: TNF-α/IL-10; I: IFN-γ/IL-4. aP < 0.05. ALC: Absolute lymphocyte count; LYM%: Lymphocyte percentage; NLR: Neutrophil-to-lymphocyte ratio.

Intra-group comparison: After the intervention, both groups of patients showed significant increases in CD3+, CD4+, CD4+/CD8+ ratio, ALC, LYM%, and TNF-α/IL-4 ratio, along with a significant decrease in NLR compared to their pre-intervention levels (P < 0.05).

Comparison of nutritional status between two groups of patients

Comparison between groups: Before the intervention, there was no significant difference in the CONUT score or PA levels between the two groups (P > 0.05). After the intervention, the proportion of patients with a CONUT score > 2 in the observation group (10.71% vs 66.67%) was significantly lower than that in the control group, and the PA level (141.56 ± 37.38 mg/L vs 260.97 ± 61.84 mg/L) was also significantly lower than that in the control group (P < 0.001, Table 3).

Table 3 Between-group comparisons of nutritional status.
Group
Observation group (n = 28)
Control group (n = 27)
χ2/t
P value
CONUT, n (%)Before intervention> 2 points15 (53.57)21 (77.78)3.5620.059
≤ 2 points13 (46.43)6 (22.22)
After intervention> 2 points3 (10.71)18 (66.67)18.2320.000
≤ 2 points25 (89.29)9 (33.33)
PA (mean ± SD, mg/L)Before intervention265.28 ± 52.31265.74 ± 52.860.0320.974
After intervention141.56 ± 37.38a260.97 ± 61.84a8.7030.000

Intra-group comparison: In the observation group, the proportion of patients at nutritional risk (CONUT score > 2) significantly decreased from 53.57% to 10.71% after the intervention (P < 0.001), and the PA level significantly decreased from 265.28 ± 52.31 mg/L to 141.56 ± 37.38 mg/L (P < 0.001). In the control group, there were no significant changes in either the nutritional risk proportion (77.78% vs 66.67%) or the PA level (265.74 ± 52.86 mg/L vs 260.97 ± 61.84 mg/L) before and after the intervention (P > 0.05, Table 4). This indicates that only the treatment regimen combined with MPBD effectively improved nutritional status.

Table 4 Within-group comparisons of nutritional status.
Group
Indicator
Before intervention
After intervention
t/χ2
P value
Observation group (n = 28)CONUT > 2, n (%)15 (53.57)3 (10.71)11.790< 0.001
PA (mean ± SD, mg/L)265.28 ± 52.31141.56 ± 37.38a10.183< 0.001
Control group (n = 27)CONUT > 2, n (%)21 (77.78)18 (66.67)0.8310.362
PA (mean ± SD, mg/L)265.74 ± 52.86260.97 ± 61.84a0.3100.758
Comparison of gastrointestinal function recovery

Patients in the observation group exhibited significantly shorter times to first flatus, bowel sound recovery, and abdominal distension relief compared to those in the control group (P < 0.05). Specific data are presented in Table 5.

Table 5 Comparison of gastrointestinal function recovery time (mean ± SD).
Group
Exhaust time (hour)
Recovery time of bowel sounds (day)
Improvement time of abdominal distension (day)
Observation group (n = 28)31.97 ± 4.352.84 ± 0.573.37 ± 0.43
Control group (n = 27)45.71 ± 4.783.81 ± 0.684.31 ± 0.56
t value11.1575.7416.998
P value0.0000.0000.000
DISCUSSION

Gastric cancer remains one of the leading global causes of cancer-related mortality, with China accounting for 44.21% of worldwide cases[15]. Despite surgical intervention, the 5-year survival rate for gastric cancer patients remains disappointingly low at only 20%, indicating suboptimal treatment outcomes[16]. This underscores the urgent need for comprehensive therapeutic approaches incorporating dietary modifications, pharmacological interventions, and long-term management strategies to improve patient prognosis. TCM attributes gastric cancer pathogenesis to spleen-stomach yin deficiency and gastric qi stagnation. MPBD, a classical Chinese herbal formula, is specifically indicated for food accumulation syndrome caused by improper or excessive dietary intake, demonstrating therapeutic effects in fortifying the spleen, promoting digestion, harmonizing the stomach, and regulating spleen-stomach qi dynamics[17,18]. This study therefore investigates the impact of MPBD on postoperative immune function and nutritional status in gastric cancer patients. This randomized controlled trial evaluated the effects of MPBD on immune function, inflammatory response, nutritional status, and gastrointestinal function recovery in gastric cancer patients after surgery. The results demonstrated that, compared to chemotherapy alone, MPBD combined with chemotherapy significantly improved all the aforementioned outcome measures.

Postoperative gastric cancer patients frequently experience issues such as immunosuppression, excessive inflammatory responses, and malnutrition, which severely hinder their recovery progress and long-term prognosis. In MPBD, A. macrocephala is the principal ingredient, and M. officinalis, dried tangerine peel and P. cocos are combined to achieve the effect of strengthening the spleen and harmonizing the stomach. P. ternata is used to tonify qi and benefit qi, calm upward movement and stop vomiting, while Forsythia is used to reduce swelling and disperse nodules, clear heat and detoxify. The combination of multiple prescriptions to a certain extent has the effect of clearing heat, unblocking the fu organs, detoxifying and eliminating pathogenic factors[19]. Previous studies have shown that Atractylodes can reduce the level of the inflammatory factor TNF-α in mice with lung injury caused by PM2.5 by 60.59%[20]. Moreover, in a mouse experiment[21], it was found that P. cocos could significantly reduce the TNF-α level in mice with skin lesions induced by ultraviolet B from 13.2 ng/mL to 7.25 ng/mL, indicating that the components such as Atractylodes and P. cocos in MPBD can alleviate the inflammatory response in patients with gastric cancer. The results of this study show that the levels of inflammatory factors such as IL-6, IFN-γ, and TNF-α in the observation group were significantly lower than those in the control group, indicating that Baohe decoction has a more significant effect in regulating gastrointestinal mucosal inflammation. The reason for the analysis is that the main ingredient in the modified MPBD is A. macrocephala, among which A. macrocephala is one of its main bioactive components. A. macrocephala can inhibit the phosphorylation and degradation of IκBα, thereby suppressing the NF-κB signaling pathway and reducing the synthesis and release of inflammatory factors in the body, resulting in a decrease in the levels of IL-6 and TNF-α in the body[22]. At the same time, Atractylodes extract can also alleviate mitochondrial dysfunction in cells and relieve the damage of oxidative stress to intestinal mucosa by eliminating reactive oxygen species and enhancing the activities of superoxide dismutase and GSH-Px in the body[23]; The main active component of M. officinalis, magnolol, can inhibit the NLRP3 inflammasome, block the oligomerization of ASC and the activation of caspase-1, reduce the maturation and release of IL-1β and IL-18, thereby activating the expression of STAT3 and RORγt, and enhancing the differentiation of naive CD4+ T cells into Th17. This further regulates the Th1/Th17 immune response, prevents excessive production of IFN-γ, and alleviates Th1-dominated intestinal inflammation[24]. The synergistic action of these mechanisms may form a crucial foundation for MPBD's efficacy in alleviating systemic inflammatory responses following surgery.

The study by Dong et al[25] demonstrated that Poria polysaccharides could significantly upregulate CD8+ and CD4+ T cell expression in C57/BL6 mice by up to twofold, indicating the close relationship between P. cocos components in MPBD and immune function. The results of this study demonstrate that regarding immune function, MPBD treatment significantly increased CD3+ and CD4+ T cell levels as well as the CD4+/CD8+ ratio, while also improving ALC and LYM%. These findings indicate its positive regulatory effect on cellular immune function. It is hypothesized that pachymaran may play a key role in this process, potentially by activating the NF-κB/MAPK signaling pathway to promote T-cell proliferation, thereby increasing CD3+ and CD4+ T-cell counts[26]. Concurrently, it may modulate the Th1/Th2 and Th17/Treg immune balance axes, further ameliorating immune dysfunction in patients and reducing recurrence risk[27]. Additionally, P. ternata in the formulation may inhibit gastric cancer progression and metastasis by regulating the PI3K/AKT/STAT3 signaling pathway, upregulating E-cadherin, p53, and Bax expression, and reducing N-cadherin levels[28], providing deeper mechanistic insights into MPBD's immunomodulatory effects.

Improved nutritional status represents another significant benefit of MPBD treatment. This study found that the proportion of patients with CONUT scores > 2 decreased significantly in the observation group, demonstrating MPBD's effectiveness in reducing postoperative malnutrition risk. This effect may be attributed to the multifaceted regulatory actions of components such as Crataegus pinnatifida and Chenpi on digestion and metabolism.

Hawthorn extract modulates the AMPK pathway to maintain intestinal barrier function, triples PPARγ levels, and subsequently reduces oxidative stress. It also alleviates lipid metabolism disorders by stabilizing the gut-liver axis, further promoting plasma PA synthesis to provide nutritional support for post-gastric cancer surgery patients[29,30]. Chenpi upregulates mucus production and tight junction protein expression while inhibiting the TLR4/NF-κB signaling pathway to reduce TNF-α levels. This normalizes serum alanine aminotransferase, aspartate aminotransferase, triglycerides, and TC levels in gastric cancer patients, thereby protecting liver function, promoting anabolic processes, and maintaining systemic nutritional reserves[31].

In terms of gastrointestinal function recovery, MPBD significantly shortened the time to first flatus, bowel sound recovery, and abdominal distension relief. β-glucans from P. cocos modulate the relative abundance of Lactobacillus and Desulfovibrio, increasing short-chain fatty acid production to enhance gut health and barrier function[32,33]. P. ternata elevates superoxide dismutase activity by 46% and regulates inflammatory responses via the IL-23/JAK2/STAT3 pathway, restoring gut microbiota balance and metabolic homeostasis[34,35]. The synergistic effects of these components collectively promote the repair and functional recovery of the gastrointestinal mucosa.

CONCLUSION

In conclusion, MPBD effectively improves postoperative immune function, reduces inflammatory responses, enhances nutritional status recovery, and accelerates gastrointestinal function restoration in gastric cancer patients, providing a novel integrated traditional Chinese and Western medicine therapeutic approach for postoperative rehabilitation. However, this study has limitations including a relatively small sample size and single-center design, which may introduce bias; future research should expand the sample size and adopt multicenter collaborative designs to enhance the generalizability of the findings.

ACKNOWLEDGEMENTS

The authors would like to express their sincere gratitude to all the patients who participated in this study. We also extend our thanks to the medical and nursing staff of the Department of Gastrointestinal Surgery at Hebei Provincial People's Hospital and The Fourth Hospital of Hebei Medical University for their invaluable assistance in patient recruitment, data collection, and clinical support. We are also grateful to our colleagues from the TCM Pharmacy for their expertise in preparing the herbal decoctions. Finally, we acknowledge the statistical guidance provided by the Clinical Research Center of Hebei Provincial People's Hospital.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade C

Scientific Significance: Grade C

P-Reviewer: Mannelli L, PhD, Italy S-Editor: Lin C L-Editor: A P-Editor: Wang CH

References
1.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 75126]  [Cited by in RCA: 67235]  [Article Influence: 16808.8]  [Reference Citation Analysis (185)]
2.  Wang HM, Wang TJ, Huang CS, Liang SY, Yu CH, Lin TR, Wu KF. Nutritional Status and Related Factors in Patients with Gastric Cancer after Gastrectomy: A Cross-Sectional Study. Nutrients. 2022;14:2634.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 31]  [Reference Citation Analysis (0)]
3.  Wan GY, Zheng LY, Li HQ, Yuan H, Xue H, Zhang XY. Effects of enteral nutritional rich in n-3 polyunsaturated fatty acids on the nutritional status of gastrointestinal cancer patients: a systematic review and meta-analysis. Eur J Clin Nutr. 2020;74:220-230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
4.  Chen M, Liang H, Chen M, Wang M, Lin L, Zhou C, Wei L. Risk factors for surgical site infection in patients with gastric cancer: A meta-analysis. Int Wound J. 2023;20:3884-3897.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 18]  [Reference Citation Analysis (0)]
5.  Lv AQ, Huang LC, Lao WL, Song QL, Zhou QF, Jiang ZM, Chen ZH. Effects of different depth of anesthesia on perioperative inflammatory reaction and hospital outcomes in elderly patients undergoing laparoscopic radical gastrectomy. BMC Anesthesiol. 2022;22:328.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
6.  Maharjan U, Kauppila JH. Survival trends in gastric cancer patients between 1987 and 2016: a population-based cohort study in Finland. Gastric Cancer. 2022;25:989-1001.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
7.  Wang Q, Chen Y, Peng Y, Yuan H, Chen Z, Wang J, Xue H, Zhang X. Preferences of oral nutritional supplement therapy among postoperative patients with gastric cancer: Attributes development for a discrete choice experiment. PLoS One. 2022;17:e0275209.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
8.  Xu W, Li B, Xu M, Yang T, Hao X. Traditional Chinese medicine for precancerous lesions of gastric cancer: A review. Biomed Pharmacother. 2022;146:112542.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 100]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
9.  Hu Y, Pan X, Nie M, Liu Y, Zou X, Liu S, Liu Q, Wang R, Zhang L. A clinical study of Yiqi Huayu Jiedu decoction reducing the risk of postoperative gastric cancer recurrence and metastasis: Study protocol for a randomized controlled trail. Medicine (Baltimore). 2020;99:e21775.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
10.  Zhang Y, Wang X, Yang H. Effect of traditional Chinese medicine nursing on postoperative patients with gastric cancer and its impact on quality of life. Am J Transl Res. 2021;13:5589-5595.  [PubMed]  [DOI]
11.  Guo K, Yan Y, Zeng C, Shen L, He Y, Tan Z. Study on Baohe Pills Regulating Intestinal Microecology and Treating Diarrhea of High-Fat and High-Protein Diet Mice. Biomed Res Int. 2022;2022:6891179.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
12.  Yasuda K. Early gastric cancer: diagnosis, treatment techniques and outcomes. Eur J Gastroenterol Hepatol. 2006;18:839-845.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
13.  Noh SH, Park SR, Yang HK, Chung HC, Chung IJ, Kim SW, Kim HH, Choi JH, Kim HK, Yu W, Lee JI, Shin DB, Ji J, Chen JS, Lim Y, Ha S, Bang YJ; CLASSIC trial investigators. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:1389-1396.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 783]  [Cited by in RCA: 788]  [Article Influence: 71.6]  [Reference Citation Analysis (0)]
14.  Ignacio de Ulíbarri J, González-Madroño A, de Villar NG, González P, González B, Mancha A, Rodríguez F, Fernández G. CONUT: a tool for controlling nutritional status. First validation in a hospital population. Nutr Hosp. 2005;20:38-45.  [PubMed]  [DOI]
15.  Huang Y, Shao Y, Yu X, Chen C, Guo J, Ye G. Global progress and future prospects of early gastric cancer screening. J Cancer. 2024;15:3045-3064.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 27]  [Reference Citation Analysis (0)]
16.  Liu Y, Huang T, Wang L, Wang Y, Liu Y, Bai J, Wen X, Li Y, Long K, Zhang H. Traditional Chinese Medicine in the treatment of chronic atrophic gastritis, precancerous lesions and gastric cancer. J Ethnopharmacol. 2025;337:118812.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
17.  Zhou X, Zhang C, Shen W, Zhuang R, Liu S, Liu J. Curzerene suppresses epithelial-mesenchymal transition in gastric precancerous lesion cells through targeted regulation of YAP via the long non-coding RNA AFAP1-AS1. Phytomedicine. 2025;143:156879.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
18.  Zhou K, Yi X, Tan Z, Peng M, Xiao N. Baohe pill decoction treats diarrhea induced by high-fat and high-protein diet by regulating lactase-producing bacteria in intestinal mucosa. Front Microbiol. 2023;14:1157475.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
19.  Zhou K, Deng N, Yi X, Cai Y, Peng M, Xiao N. Baohe pill decoction for diarrhea induced by high-fat and high-protein diet is associated with the structure of lactase-producing bacterial community. Front Cell Infect Microbiol. 2022;12:1004845.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
20.  Yun EH, Bashir KMI, Lee J, Chung H, Kwon YS, Choi JS, Ku SK. Protective Effects of Atractylodis Rhizoma Extracts on Lung Injury Induced by Particulate Matter 2.5 in Mice. Antioxidants (Basel). 2025;14:127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
21.  Zhang T, Huang S, Qiu J, Wu X, Yuan H, Park S. Beneficial Effect of Gastrodia elata Blume and Poria cocos Wolf Administration on Acute UVB Irradiation by Alleviating Inflammation through Promoting the Gut-Skin Axis. Int J Mol Sci. 2022;23:10833.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
22.  Shi K, Xiao Y, Qu M, Xie Y, Wang Y, Ke C, Qu L, Liu Y. Atractylodin modulates ASAH3L to improve galactose metabolism and inflammation to alleviate acute lung injury. iScience. 2024;27:110751.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
23.  Li Z, Song Y, Hou W, Qi Y, Lu X, Xue Y, Huang J, Fang Q. Atractylodin induces oxidative stress-mediated apoptosis and autophagy in human breast cancer MCF-7 cells through inhibition of the P13K/Akt/mTOR pathway. J Biochem Mol Toxicol. 2022;36:e23081.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
24.  Tao L, Zhang Q, Liu L, Wang K, Liu X, Li J, Zhao P. Magnolol preserves the integrity of the intestinal epithelial barrier and mitigates intestinal injury through activation of PPAR γ in COPD rat. J Ethnopharmacol. 2025;338:119068.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
25.  Dong X, Li B, Yu B, Chen T, Hu Q, Peng B, Sheng W. Poria cocos polysaccharide induced Th1-type immune responses to ovalbumin in mice. PLoS One. 2021;16:e0245207.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
26.  Zhang W, He J, Zheng D, Zhao P, Wang Y, Zhao J, Li P. Immunomodulatory Activity and Its Mechanisms of Two Polysaccharides from Poria cocos. Molecules. 2023;29:50.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
27.  Tu Y, Luo X, Liu D, Li H, Xia H, Ma C, Zhang D, Yang Y, Pan X, Wang T, Xia Y, Dan H, You P, Ye X. Extracts of Poria cocos improve functional dyspepsia via regulating brain-gut peptides, immunity and repairing of gastrointestinal mucosa. Phytomedicine. 2022;95:153875.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 27]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
28.  Duan ZW, Liu Y, Zhang PP, Hu JY, Mo ZX, Liu WQ, Ma X, Zhou XH, Wang XH, Hu XH, Wei SL. Da-Chai-Hu-Tang Formula inhibits the progression and metastasis in HepG2 cells through modulation of the PI3K/AKT/STAT3-induced cell cycle arrest and apoptosis. J Ethnopharmacol. 2024;331:118293.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
29.  Han X, Zhao W, Zhou Q, Chen H, Yuan J, Xiaofu Z, Zhang Z. Procyanidins from hawthorn (Crataegus pinnatifida) alleviate lipid metabolism disorder via inhibiting insulin resistance and oxidative stress, normalizing the gut microbiota structure and intestinal barrier, and further suppressing hepatic inflammation and lipid accumulation. Food Funct. 2022;13:7901-7917.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
30.  Li S, Liu Y. Intestinal absorption mechanism and nutritional synergy promotion strategy of dietary flavonoids: transintestinal epithelial pathway mediated by intestinal transport proteins. Crit Rev Food Sci Nutr. 2025;65:4368-4381.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
31.  Zhang XM, Huang YC, Chen BZ, Li Q, Wu PP, Chen WH, Wu RH, Li C. Water decoction of Pericarpium citri reticulatae and Amomi fructus ameliorates alcohol-induced liver disease involved in the modulation of gut microbiota and TLR4/NF-κB pathway. Front Pharmacol. 2024;15:1392338.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
32.  Li JH, Gu FT, Yang Y, Zhao ZC, Huang LX, Zhu YY, Chen S, Wu JY. Simulated human digestion and fermentation of a high-molecular weight polysaccharide from Lentinula edodes mushroom and protective effects on intestinal barrier. Carbohydr Polym. 2024;343:122478.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 17]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
33.  Liu Z, Tang R, Liu J, Zhang Z, Li Y, Zhao R. Epicatechin and β-glucan from whole highland barley grain ameliorates hyperlipidemia associated with attenuating intestinal barrier dysfunction and modulating gut microbiota in high-fat-diet-fed mice. Int J Biol Macromol. 2024;278:134917.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
34.  Wang J, Chen J, Zhang X, Feng X, Li X. Physiological and transcriptional responses to heat stress in a typical phenotype of Pinellia ternata. Chin J Nat Med. 2023;21:243-252.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
35.  Yang Y, Guo L, Yu J, Peng W, Wang Q, Wei L, Chen X, Min C, Jiang X, Zhu S, Zhao H, Xuan Z, Yu J, Zhang J. Da-Yuan-Yin decoction suppresses NETs formation, inhibits the IL-23/JAK2/STAT3 signalling pathway, and modulates metabolic profiles and gut microbiota composition in damp-heat syndrome. Ann Med. 2025;57:2519684.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]