China Oncology
Co-Editors-in-Chief
2025
Volume: 35 Issue 7
11 Article(s)
PENG Peng, DOU Jianming, WU Chunxiao, PANG Yi, GONG Yangming, WU Mengyin, SHI Yan, and GU Kai

Background and purposeThe colorectal cancer screening program for community residents in Shanghai has been implemented for 12 years since 2013. This study aimed to analyze the impact of screening on the colorectal cancer incidence, stage and survival outcomes based on their screening participation status.MethodsThis study used registry-based cohort study method. The registered residents in Shanghai from 2013 to 2017 who met the screening age range were divided into screening group and non-screening group. The data of colorectal cancer cases after being included in groups were obtained from the Population Based Cancer Registry. We calculated age-standardized cumulative incidence and age-group cumulative incidence, diagnosis stage and survival rate of colorectal cancer by gender, age and year of diagnosis. We used the Joinpoint regression method to calculate the annual change percentage for cumulative incidence trend analysis. The life table method and Ederer Ⅱ method were used to calculate the 5-year observed survival rates and expected survival rates of colorectal cancer cases. Finally the 5-year relative survival rates were obtained.ResultsThe study included 1 687 689 participants aged 50-74 in screening group and 4 713 307 participants in non-screening group. During a 5-year follow-up period, there were 10 333 and 20 904 new cases of colorectal cancer diagnosed in the two groups, respectively. The age-standardized 5-year cumulative incidence in the screening group was 555.33/105, with an average annual increase of 33.32% (P<0.05). The age-standardized 5-year cumulative incidence in the non-screening group was 529.85/105, with an average annual increase of 48.13% (P<0.05). There was no statistically significant difference between the screening group and the non-screening group in the age-standardized 5-year cumulative incidence (X=0.25, P=0.804). The lower the age group, the greater the difference between the screening group and the non-screening group in the annual average change percentage of the age-standardized cumulative incidence. The stages 0-Ⅰ of newly diagnosed colorectal cancer cases in the screening group and non-screening group accounted for 14.70% and 7.46%, respectively, with a statistically significant difference in composition between the two groups (P<0.05). The 5-year relative survival rate of the screening group was 73.94%, while the non-screening group was 59.66%. The survival rate indicators of the former were significantly higher than those of the latter, and the difference was statistically significant. The survival rate of the former was significantly higher than that of the latter (73.94% vs 59.66%), and the difference was statistically significant (P<0.05). The survival rate of females in both groups of cases was higher than that of males, and the survival rate decreased with increasing age-groups at diagnosis.ConclusionWith the implementation of the colorectal cancer screening program, the growth trend of the incidence rate of colorectal cancer among the screening participants has been curbed, and the early stages of colorectal cancer cases diagnosed and the 5-year survival rate were significantly improved. In order to reduce the incidence rate of colorectal cancer in the whole population, it is necessary to vigorously promote the screening coverage of the appropriate population, especially to increase the proportion of lower age groups participating in screening. We should also pay attention to the screening quality of the elderly groups and improve the compliance of colonoscopy in high-risk participants. At the same time, we should further optimize the refined management of screening for different genders, ages, and risk groups.

Aug. 22, 2025
  • Vol. 35 Issue 7 657 (2025)
  • AN Yang, WANG Chentong, QIU Xiaoyuan, ZHOU Jiaolin, and LIN Guole

    Background and PurposeNeoadjuvant immunotherapy currently significantly enhances treatment efficacy for locally advanced rectal cancer (LARC); However, clinical management of immune-related adverse events (irAEs) lacks robust evidence. This study aimed to investigate the characteristics, clinical management strategies, and outcomes of irAEs during neoadjuvant immunotherapy for rectal cancer, providing a basis for optimizing irAEs monitoring and intervention.MethodsWe retrospectively analyzed clinical data from LARC patients who developed irAEs after receiving neoadjuvant immunotherapy at Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, between July 2022 and June 2024. Types of irAEs, severity, time of onset, management strategies, and outcomes were recorded. All patients underwent regular follow-up for at least 6 months. This study has been approved by Peking Union Medical College Hospital, Chinese Academy of Medical Sciences (ethical approval number: I-24PJ0024). Descriptive statistics were used to summarize irAEs patterns and management approaches.ResultsA total of 41 irAE episodes occurred among the 30 patients. Mild irAEs (Grade 1-2) accounted for 78.0% (32/41), while severe irAEs (Grade 3-4) constituted 22.0% (9/41). Five patients (16.7%) permanently discontinued treatment due to severe toxicity. Endocrine toxicities were most frequent (36.6%, 15/41), primarily characterized by progression from hyperthyroidism to hypothyroidism; 75.0% required thyroid hormone replacement therapy. One case of delayed-onset adrenal insufficiency was alleviated with glucocorticoid (GC) therapy. Among hepatotoxicities (19.5%, 8/41), 62.5% were Grade 3 injury, and 37.5% required GC intervention; two patients experienced recurrence during adjuvant chemotherapy. Three cases of severe myositis occurred, accompanied by asymptomatic myocardial injury (evidenced by markedly elevated creatine kinase and concurrent changes in cardiac biomarkers), all requiring high-dose GC pulse therapy combined with intravenous immunoglobulin or immunosuppressants (recovery period: 2-4 months). Nine dermatological reactions were managed with topical therapy. Two gastrointestinal events occurred, including one Grade 3 diarrhea treated with GCs. The overall GC usage rate was 31.7% (13/41), with 76.9% administered for Grade ≥3 irAEs.ConclusionirAEs during neoadjuvant immunotherapy for LARC are predominantly mild-to-moderate and manageable with supportive care. However, some patients develop severe (Grade 3-4) irAEs requiring multidisciplinary management. GC usage is concentrated in highergrade irAEs, with severe myositis and cardiac involvement necessitating intensive immunosuppressive therapy despite their rarity. Recurrence of irAEs during adjuvant chemotherapy in a minority of patients underscores the necessity for early recognition, graded intervention, and comprehensive management throughout the entire treatment cycle.

    Aug. 22, 2025
  • Vol. 35 Issue 7 665 (2025)
  • LIU Shuo, LU Yun, HU Jilin, YANG Wenchang, ZHAO Rizhi, XU Wenda, YANG Hanyu, LU Zechen, MA Zheng, DU Zhaolin, GAO Yunzhi, and GAO Yuan

    Background and PurposePrevious studies have investigated the prognostic significance of skeletal muscle and adipose tissue composition and distribution in colorectal cancer patients, yet most have not differentiated between rectal and colon cancer patient cohorts. This study aimed to explore the relationship between body composition and long-term prognosis, and to develop a postoperative predictive model.MethodsClinical data of rectal cancer patients who underwent surgical treatment at Qingdao University Affiliated Hospital from January 2018 to December 2021 were retrospectively collected. Inclusion criteria: ①Age>18 years; ② Preoperative colonoscopy and pathological diagnosis of colorectal cancer; ③ Complete surgical resection; ④Abdominal computed tomography (CT) scan 1 month before surgery. Exclusion criteria: ① Clinical data is missing; ② Multiple metastases of tumors; ③ Tumor T stage 0 or carcinoma in situ; ④ Severe artifacts lead to poor quality CT imaging, making it difficult to distinguish between fat and muscle; ⑤ Inability to obtain follow-up results. This study has been approved by the Medical Ethics Committee of the Affiliated Hospital of Qingdao University (approval number: QYFYWZLL30313), and informed consent has been waived in the ethical approval process. The skeletal muscle index (SMI) and subcutaneous adipose tissue index (SATI) were calculated by dividing the areas of skeletal muscle and subcutaneous fat observed on CT scans by the square of the patient's height. Univariate and multivariate COX regression analyses were conducted to identify risk factors influencing recurrence-free survival (RFS) and overall survival (OS) in rectal cancer patients. Based on the results of the multivariate analysis, a nomogram prediction model was developed, its predictive power and accuracy were assessed using the receiver operating characteristic (ROC) curve, calibration plots and decision curve analysis (DCA), and internal validation was conducted.ResultsA total of 696 patients were included in this study, with 96 (13.8%) patients experiencing postoperative recurrence and 89 (12.8%) patients dying. Multivariate COX regression analysis showed that SMI, SATI, tumor T stage and N stage were independent factors affecting the postoperative RFS and OS of patients. Nomogram prediction models for RFS and OS in rectal cancer patients were constructed based on the above independent predictors. The area under ROC curve (AUC) for 3-, 4- and 5-year RFS was 0.862, 0.846 and 0.824, respectively; the AUC for 3-, 4- and 5-year OS was 0.886, 0.898 and 0.875, respectively. The models were evaluated using calibration curves and decision curves, and internal validation was performed, which showed that the prediction accuracy of the models was good.ConclusionCT body composition is an independent predictor of RFS and OS in rectal cancer patients, and the nomogram model developed based on these factors demonstrates good predictive value for patient prognosis.

    Aug. 22, 2025
  • Vol. 35 Issue 7 672 (2025)
  • Aug. 22, 2025
  • Vol. 35 Issue 7 1 (2025)
  • LI Xinxiang, and LUO Dakui

    In recent years, the surgical treatment model for rectal cancer has undergone profound changes. The therapeutic goal has gradually shifted from single tumor radical resection to balancing functional preservation, and the therapeutic concept has transformed from merely emphasizing surgical techniques to attaching importance to comprehensive treatment. Especially in the treatment of low rectal cancer, the neoadjuvant therapy model has been continuously optimized. For patients with good tumor regression after neoadjuvant therapy, “watch and wait” and transanal local excision have become important optional strategies. This not only avoids some severe surgery-related complications but also maximizes the preservation of patients' organ functions, bringing a qualitative leap in their quality of life. This treatment strategy is gradually expanding from locally advanced low rectal cancer to relatively early-stage low rectal cancer. In terms of surgical techniques, based on the traditional intermediate approach of “first plane, then vessels”, the concept of a “vessel-centered” approach is proposed. By managing vessels first and then expanding the plane, it enables thorough dissection of lymph nodes at the root of the inferior mesenteric artery while preserving the left colic artery. With the aid of dual-fluorescence intraoperative navigation technology [indocyanine green (ICG) fluorescence and intraoperative realtime imaging system (IRIS) ureter fluorescence imaging], real-time visualization of lymph nodes and ureters is achieved, ensuring the completeness of lymph node dissection and helping to reduce the risk of ureteral injury. The angulation-free double anastomosis technique used during surgery effectively reduces the incidence of anastomotic leakage and improves surgical safety. For patients with high-risk factors for anastomotic leakage, intestinal stent bypass is expected to replace the traditional prophylactic end ileostomy, thus avoiding complications associated with prophylactic end ileostomy and the trauma caused by secondary stoma closure. In general, the development trend of surgical treatment for rectal cancer is to minimize patient trauma, preserve organ functions, and improve quality of life under the premise of ensuring oncological efficacy, promoting the development of surgical techniques towards standardization and precision to maximize patients' perioperative safety.

    Aug. 22, 2025
  • Vol. 35 Issue 7 631 (2025)
  • WEI Pengyu, GAO Jiale, JI Maotong, TAN Yuanyuan, and YAO Hongwei

    Laparoscopic right hemicolectomy is the standard surgical procedure for treating right-sided colon cancer, yet its complex anatomical layers and technically demanding operations pose significant challenges to surgeons' expertise. Currently, surgical skill assessment in clinical practice still predominantly relies on subjective expert evaluation, an approach inherently limited by low efficiency and inconsistent standards. With the rapid advancement of artificial intelligence (AI) technologies (particularly breakthroughs in computer vision and deep learning algorithms), a revolutionary technical foundation has been established for developing objective and precise automated surgical evaluation systems. This article comprehensively reviewed the current clinical application status of right hemicolectomy in China, thoroughly examined the clinical necessity and technical feasibility of constructing an intelligent surgical assessment system, and systematically presented our center's cutting-edge research achievements in this field. Looking ahead, AI-driven intelligent evaluation systems are expected to standardize and quantify surgical skill assessment. Such innovation will not only transform surgical training programs and enhance the quality of multicenter clinical research, but also promote the standardization of precision surgical protocols. This advancement holds significant clinical value and societal importance by improving long-term patient outcomes and fostering more equitable distribution of medical resources.

    Aug. 22, 2025
  • Vol. 35 Issue 7 637 (2025)
  • ZHANG Yuyang, and LIU Qian

    In recent years, significant progress has been made in neoadjuvant immunotherapy for colorectal cancer (CRC), particularly demonstrating breakthrough efficacy in mismatch repair deficient/microsatellite instability-high (dMMR/MSI-H) subtypes. Immune checkpoint inhibitor (ICI) has achieved complete response (CR) rates as high as 41%-100% in this patient subgroup, driving the clinical adoption of “surgery-sparing” and “watch-and-wait” strategies. However, the majority of CRC patients with mismatch repair proficient/microsatellite stable (pMMR/MSS) tumors show limited response to ICI monotherapy, necessitating combination approaches with radiotherapy, chemotherapy, or targeted therapies to enhance efficacy. Current studies indicate that such combined regimens can elevate pathological complete response (pCR) rates to 22%-63%. Key research focuses include the synergistic effects of short-course radiotherapy (SCRT) combined with immunotherapy, the potential of dual-ICI therapy, and precision patient selection using biomarkers such as POLE/POLD1 mutations and tumor mutational burden (TMB). For treatment response assessment, the integration of colonoscopy, imaging, circulating tumor DNA (ctDNA) and artificial intelligence (AI) holds promise for optimizing clinical decision-making. Future efforts should prioritize immunomodulation strategies for pMMR/MSS patients, long-term safety evaluation of organ preservation approaches, and multidisciplinary collaboration to advance personalized therapy. Neoadjuvant immunotherapy is reshaping the CRC treatment paradigm, offering improved survival and quality of life for patients.

    Aug. 22, 2025
  • Vol. 35 Issue 7 642 (2025)
  • LIU Dan, ZHANG Guxiang, XIE Dan, XU Yan, XIE Chengfang, and TANG Yuxi

    Background and purposeImmune checkpoint blockade (ICB) has become a promising strategy for treating cervical cancer (CC), but terminal T cell depletion still limits the therapeutic efficacy of ICB. The deletion of sorting nexin-9 (SNX9) can inhibit thymocyte selection-associated high mobility group box protein (TOX), alleviate T cell exhaustion, and provide new ideas for preventing T cell exhaustion and enhancing the efficacy of cancer immunotherapy. Therefore, this study aimed to explore the immune escape mechanism mediated by the depletion of CD8+ T cells induced by the SNX9/TOX signaling pathway in the CC microenvironment.MethodsFifty-four peripheral blood samples were collected, including 18 from CC patients, 18 from patients with high-grade squamous intraepithelial lesions (HSIL) and 18 from subjects with normal cervix. In addition, the study collected 153 pairs of CC and adjacent tissues from patients who received operation in our hospital for the first time. Clinicopathological features, tumor stages, follow-up records and other relevant clinicopathological data of CC patients were obtained from hospital records. The research was approved by the ethics committee of Changsha Fourth Hospital (approval number: 20220206). A total of 24 mice were randomly assigned to the following four groups: immunoglobulin G (IgG) group, Anti-SNX9 group, Anti-programmed death-1 (PD-1) group and Anti-SNX9+ Anti-PD-1 group, with 6 mice in each group. Each group received intraperitoneal injection of blocking antibody and isotype control treatment respectively. ELISpot was used to detect the ability of CD8+ T cells to secrete tumor necrosis factor- (TNF-) and interferon- (IFN-). The expressions of TOX and SNX9 in cervical cancer tissues were detected by western blot and immunohistochemistry.ResultsThe expressions of SNX9 and TOX mRNA in peripheral blood mononuclear cell (PBMC) of CC patients were higher compared with HSIL and normal controls (P<0.05). The positive cell level of SNX9 and TOX immunohistochemical score were higher in CC tissue than in adjacent tissues (t=18.63, 21.10, P<0.001). The high expression of SNX9 in CC was related to low differentiation/undifferentiation, tumor size, parauterine infiltration, vaginal infiltration, late FIGO stage and pelvic lymph node metastasis (P<0.05). Compared with the low expression group of SNX9, the overall survival time of CC patients in the high expression group of SNX9 was shorter (P<0.05). The percentage of CD8+ SNX9+ T cells was significantly higher in CC patients than in normal controls and HSIL patients (P<0.05). The ability of CD8+ SNX9+ T cells to secrete cytokines (TNF- and IFN-) was significantly lower compared with CD8+ SNX9- T cells (P<0.05). Compared with the Anti-SNX9 group, the growth and proliferation of cervical tumor, the expression of SNX9 and TOX protein in tumor tissue in the Anti-SNX9+Anti-PD-1 group further decreased (P<0.05), and the level of infiltrating CD8+ T lymphocytes in tumor tissue and the ability of CD8+ T lymphocytes to secrete functional factors TNF- and IFN- further increased (P<0.05).ConclusionSNX9/TOX signaling pathway exhibits enhanced activity in patients with cervical cancer and mouse models, and is related to immunosuppression. Targeting SNX9/TOX signaling pathway may be a potential therapeutic strategy for CC.

    Aug. 22, 2025
  • Vol. 35 Issue 7 685 (2025)
  • XIANG Shiyu, LI Qiao, SHEN Changqing, GU Yajia, and WU Bin

    Background and purposeAccurate preoperative differentiation between vagal nerve cervical schwannomas (SCCS) and sympathetic chain cervical schwannomas (SCCS) in the neck is crucial because of their different postoperative complication. This study aimed to construct and validate a Diagnosis Score and vascular displacement nomogram for the preoperative differentiation of VNCS from SCCS in the neck.MethodsThis cross-sectional study retrospectively analyzed patients with pathologically confirmed VNCS and SCCS at Fudan University Shanghai Cancer Center from January 2017 to April 2022. This study was approved by the medical ethics committee of Fudan University Shanghai Cancer Center (approval number: 1612167-18). Inclusion criteria: ① histopathological diagnosis of VNCS or SCCS through biopsy or surgical resection; ② patients with complete clinic data; ③availability of preoperative contrast-enhanced computed tomography (CT) or magnetic resonance imaging (MRI) examinations. Patients were excluded for: ① contrast agent contraindications; ② poor image quality; ③ severe artifacts; ④ non-standard scanning protocols. The cohort was randomly divided into training and validation sets in a 7∶3 ratio. Two radiologists (one resident and one attending physician) independently evaluated tumor characteristics (location, size and vascular displacement patterns) on preoperative imaging. Independent predictors were selected using LASSO regression analysis to construct a diagnostic scoring system and nomogram, with model performance evaluated by the receiver operating characteristic (ROC) curve.ResultsA total of 110 patients were enrolled, with 77 cases allocated to the training set and 33 cases to the validation set. The age range was 24-78 years, and the mean age was (51.22±12.36) years. There were no statistically significant differences in baseline characteristics including age, gender, tumor location and size between the two patient groups (P>0.05). ICA/ECA splaying was significantly associated with SCCS (P<0.001), while the ICA/IJV splaying was significantly associated with VNCS (P<0.001). Lateral and posterior ICA displacement were significantly associated with SCCS (P<0.001), and medial and anterior ICA displacement were significantly associated with VNCS (P<0.001). Five features including tumor size, ICA displacement direction, IJV displacement direction, ICA/ECA splaying and ICA/IJV splaying were used to establish the Diagnosis Score and nomogram. The nomogram combined imaging features showed favorable preference value for differentiating VNCS from SCCS, with area under curve (AUC) values of 0.953 (95% CI: 0.912-0.994) and 0.939 (95% CI: 0.885-0.993) for the training and validation cohorts, respectively.ConclusionThe Diagnosis Score and vascular displacement nomogram showed favorable predictive efficacy for differentiating VNCS from SCCS in the neck, and might be useful for clinical decision-making.

    Aug. 22, 2025
  • Vol. 35 Issue 7 695 (2025)
  • QIAN Jiajia, RUAN Cong, LIU Jiyong, and XU Rui

    In recent years, immune checkpoint inhibitor (ICI) has led to substantial advances in the treatment of recurrent or metastatic advanced cutaneous melanoma (CM), significantly prolonging overall survival. However, due to the biological heterogeneity across melanoma subtypes, the degree of immune responsiveness varies considerably. In particular, acral melanoma (AM) (the predominant melanoma subtype in Asian populations, including China) has demonstrated limited benefit from ICI therapy, especially in the context of monotherapy. Currently, no systematic staging and standardized treatment guidelines are available for AM, and clinical evidence supporting the use of ICI in this rare subtype remains insufficient. In the neoadjuvant setting, several large phase Ⅱ/Ⅲ international trials in CM, including SWOG 1801 and NADINA, have shown that ICI-based neoadjuvant combination therapy significantly improves pathological response rates compared with traditional adjuvant approaches. Nevertheless, neoadjuvant treatment in AM remains in the exploratory stage. Early-phase clinical studies in resectable stage Ⅲ/Ⅳ AM suggest that toripalimab combined with intratumoral oncolytic virus therapy, or camrelizumab in combination with apatinib and temozolomide, may offer clinical benefit; however, confirmation of long-term survival benefit requires further validation in larger, prospective cohorts. In the adjuvant setting, for AM patients with BRAF mutations, real-world data from China have shown no significant difference in survival outcomes between dabrafenib plus trametinib and programmed death-1 (PD-1) inhibitor monotherapy in high-risk resectable stage Ⅲ/Ⅳ disease, although direct head-to-head comparisons are lacking. For patients with resectable stage Ⅲ/Ⅳ wild-type AM, combination adjuvant regimens incorporating PD-1 inhibitors may provide superior recurrence risk reduction and survival benefit compared to monotherapy. In the advanced disease setting, in Chinese populations, the objective response rates of PD-1 inhibitors such as pembrolizumab, toripalimab and penpulimab remain suboptimal in AM. ICI-based combination strategies (including those with chemotherapy, anti-angiogenic agents, dual or triple immune checkpoint blockade) may improve the immune microenvironment and clinical prognosis, but concerns regarding safety and tolerability persist. For patients with ICI-refractory AM, various novel approaches combining immunotherapy, targeted agents and chemotherapy are under investigation. Additionally, several nextgeneration immunotherapeutic modalities [including T-cell receptor-engineered (TCR-T) therapies, therapeutic cancer vaccines, chimeric antigen receptor T (CAR-T) cell therapy and antibody-drug conjugate (ADC)] are currently in development. This review aimed to provide a comprehensive overview of current clinical evidence on the use of ICI in acral melanoma across the neoadjuvant, adjuvant, and advanced disease settings. We highlighted the efficacy and safety of existing strategies, exploreed emerging combination regimens and predictive biomarkers, and discussed key areas for future research to inform clinical decision-making and optimize outcomes in this challenging melanoma subtype.

    Aug. 22, 2025
  • Vol. 35 Issue 7 702 (2025)
  • WANG Hongxia, YIN Yongmei, and HU Xichun

    Breast cancer remains one of the frequently diagnosed malignant tumors among Chinese women, with hereditary cases accounting for 5%-10% of all diagnoses, where BRCA1/2 gene mutations serve as the primary genetic predisposition factors. Although targeted therapies like poly (ADP-ribose) polymerase (PARP) inhibitors have significantly improved prognoses for patients with BRCA-mutated breast cancer in recent years, critical clinical challenges persist, including the standardization of genetic testing protocols, optimization of precision treatment approaches, and refinement of long-term management strategies. In response to these challenges, our expert panel has conducted a comprehensive update to the 2018 Edition of this consensus by integrating the latest global evidence-based medical research with China's unique clinical practice characteristics. This 2025 Edition provides systematic evaluations and recommendations on five key aspects: indications for BRCA1/2 gene testing, testing methodologies, result interpretation, treatment strategies, and risk management. The main updates include: ① Increasing the relationship between BRCA1/2 gene mutations and programmed death ligand-1 (PD-L1) expression, as well as related content on BRCAness types; ② Standardizing the application of genetic testing, such as increasing the significance, timing, and sample selection of clinical testing, and optimizing the BRCA testing population; ③ Updating treatment strategies, such as non-drug treatment of BRCA1/2 gene mutation, treatment of triple negative breast cancer (TNBC) patients with BRCA1/2 gene mutation, treatment decisions of hormone receptor (HR)+/human epidermal growth factor receptor 2 (HER2)- breast cancer patients with BRCA1/2 gene mutation, clinical use of PARP inhibitors and adverse reaction management; ④ Addion of relevant content on long-term risk management, such as covering follow-up management, indications for preventive surgery, quality control and requirements for new genetic testing, updating genetic testing processes, report content and interpretation. This consensus aimed to establish standardized diagnostic and therapeutic frameworks for clinicians, advance precision medicine in BRCA-mutated breast cancer, and ultimately improve patient survival outcomes. As new evidence emerges, continuous updates will be implemented to incorporate the latest research findings. This consensus has been registered on the Practice guideline REgistration for transPAREncy (PREPARE) platform (registration number: PREPARE-2025CN1085).

    Aug. 22, 2025
  • Vol. 35 Issue 7 710 (2025)
  • Please enter the answer below before you can view the full text.
    Submit