Retrospective Study Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. May 21, 2025; 31(19): 106644
Published online May 21, 2025. doi: 10.3748/wjg.v31.i19.106644
Pescadillo ribosomal biogenesis factor 1 and programmed death-ligand 1 in gastric and head and neck squamous cell carcinoma
Xiao-Nan Hu, Department of Head and Neck Radiotherapy Ward, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
Chun-Feng Li, Department of Gastrointestinal Surgical Ward, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
Si-Meng Huang, Chun-Lei Nie, Rui Pang, Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
ORCID number: Rui Pang (0000-0001-9015-9290).
Co-first authors: Xiao-Nan Hu and Chun-Lei Nie.
Author contributions: Hu XN, Li CF, Huang SM, and Nie CL contributed to the research design, data collection, data analysis, and paper writing; Pang R was responsible for the research design, funding application, data analysis, reviewing and editing, communication coordination, ethical review, copyright and licensing, and follow-up; All authors read and approved the final manuscript. Hu XN and Nie CL contributed equally to this work as co-first authors.
Institutional review board statement: The research was reviewed and approved by the Affiliated Cancer Hospital of Harbin Medical University.
Informed consent statement: All research participants or their legal guardians provided written informed consent prior to study registration.
Conflict-of-interest statement: No conflicts of interest were associated with this work.
Data sharing statement: No other data available.
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: Rui Pang, Deputy Director, Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Harbin 150081, Heilongjiang Province, China. 3151@hrbmu.edu.cn
Received: March 7, 2025
Revised: April 7, 2025
Accepted: April 23, 2025
Published online: May 21, 2025
Processing time: 74 Days and 20.6 Hours

Abstract
BACKGROUND

Gastric cancer (GC) and head and neck squamous cell carcinoma (HNSCC) are common malignancies with high morbidity and mortality rates. Traditional treatments often yield limited efficacy, especially in advanced cases. Recent advancements in immunotherapy, particularly immune checkpoint inhibitors targeting programmed death-ligand 1 (PD-L1), have shown promise. However, the expression and interaction of pescadillo ribosomal biogenesis factor 1 (PES1) and PD-L1 in these cancers remain unclear. Understanding their roles could provide new insights into tumor biology and improve therapeutic strategies.

AIM

To investigate the expression levels of PES1 and PD-L1 in tumor tissues of patients with GC and HNSCC.

METHODS

A total of 58 cases of GC and HNSCC undergoing surgical resection were selected from January 2022 to January 2024. Paraffin specimens of GC and HNSCC tissues were taken from the patients, and the sections were subjected to staining with immunohistochemistry and hematoxylin-eosin staining, and the protein expression of PES1 and PD-L1 was observed microscopically.

RESULTS

Among 58 GC and HNSCC tissues, 30 cases were positive and 28 cases were negative for PES1 expression, and 34 cases were positive and 24 cases were negative for PD-L1 expression. The positive expression rates of PES1 and PD-L1 were 51.72% and 58.62%, respectively. PES1 expression was correlated with the TNM stage, lymph node metastasis, and the depth of infiltration (P < 0.05), and PD-L1 expression was correlated with the differentiation degree, lymph node metastasis, and infiltration depth (P < 0.05).

CONCLUSION

PES1 and PD-L1 were positively expressed in GC and HNSCC tissues and correlated with clinical features. They may serve as potential biomarkers for immune-targeted therapies.

Key Words: Pescadillo ribosomal biogenesis factor 1; Programmed death-ligand 1; Gastric cancer; Head and neck squamous cell carcinoma; Expression level

Core Tip: This study investigated the expression of pescadillo ribosomal biogenesis factor 1 (PES1) and programmed death-ligand 1 (PD-L1) in tumor tissues from patients with gastric cancer and head and neck squamous cell carcinoma. We found that both proteins exhibited significant positive expression rates (51.72% for PES1 and 58.62% for PD-L1) and were associated with clinical features such as TNM stage and lymph node metastasis. Our results suggest that PES1 and PD-L1 may play critical roles in tumor progression and immune escape, potentially serving as biomarkers for immune-targeted therapies.



INTRODUCTION

Gastric cancer (GC) and head and neck squamous cell carcinoma (HNSCC) are common malignant tumors that pose a grave menace to human health worldwide[1,2]. GC is ranked fifth in tumor frequency across the globe and is the fifth-leading cause of cancer-related deaths, especially in China where its incidence and mortality rates remain high[3]. This scenario not only presents a significant danger to the well-being of patients but also imposes a substantial strain on the societal public health infrastructure. The high morbidity and mortality of GC requires continuous innovation and breakthroughs in treatment strategies, especially for patients with advanced and/or recurrent disease in which existing treatments, such as chemotherapy and radiotherapy, have limited efficacy.

Meanwhile, head and neck tumors are a part of the most common and widespread malignant tumors worldwide. HNSCC is the most frequently occurring head and neck tumor. The effectiveness of conventional treatments, especially in patients with advanced or recurrent disease, is still limited, and there is a pressing necessity to identify novel treatment approaches, particularly for patients who are resistant to chemotherapy or experience recurrence[4].

In recent years, with the rise of immunotherapy, especially the clinical application of immune checkpoint inhibitors, new hope has been brought to tumor treatment. Among them, programmed death-ligand 1 (PD-L1), as an important immune checkpoint molecule, is strongly expressed in a spectrum of tumor tissues and inhibits T cell activity by binding to programmed death 1 (PD-1), leading to tumor immune escape[5,6]. The PD-L1/PD-1 pathway is a cornerstone of regulating immune response and maintaining immune tolerance, and its inhibitors can block this pathway and restore T cell activity, thus enhancing immune response and inhibiting tumor growth.

PD-L1 has been demonstrated to be highly elevated in GC and HNSCC, with its expression level being significantly in line with clinicopathological features and patient outcomes. In GC, the expression of PD-L1 was closely linked to the depth of infiltration of tumor cells, the degree of differentiation, and the survival of patients. In HNSCC, PD-L1 expression was also closely in line with clinical stage. Additionally, patients with PD-L1 positivity had a higher likelihood of deriving benefits from anti-PD-1/PD-L1 treatment[7-10]. Although the important role of PD-L1 in tumor immunotherapy has been widely recognized, much remains unknown about its expression mechanism and interaction with other immune molecules.

Pescadillo ribosomal biogenesis factor 1 (PES1), as a new biomarker, has not yet been intensively investigated in terms of its expression in tumor tissues and its relationship with PD-L1[11]. Therefore, the purpose of this study was to detect the expression of PES1 and PD-L1 in tumor tissues of patients with GC and HNSCC, to explore the differences in their expression in different pathological grades and clinical stages, to provide theoretical and experimental bases for the immune-targeted therapy of GC and HNSCC, and to provide new ideas for the future immunotherapeutic strategies.

MATERIALS AND METHODS
Patient selection

A total of 58 cases of GC and HNSCC admitted to our hospital for surgical resection between January 2022 and January 2024 were selected, all of which were definitively diagnosed as GC or HNSCC by pathology. Among them, there were 37 male patients and 21 female patients. The age range was 39 to 72 years with a mean of 52.33 ± 8.29 years. Surgical resection specimens were used for analysis, and other relevant clinical data were taken from patients’ pathology reports and medical records. The utilization of the specimens for clinical purposes in this study was examined and sanctioned by the hospital’s Ethics Committee.

Inclusion criteria were: (1) Patients and their families have signed informed consent; (2) Patients have not received radiotherapy, chemotherapy, or immunotherapy for tumors; and (3) All specimens were fixed in 40 g/L formaldehyde and embedded in paraffin within 60 min after removal from the body, and the thickness of the tissue sections was 4 μm. Exclusion criteria were: (1) Patients with metastatic tumors or recurrent tumors; (2) Patients with severe consciousness disorders; (3) Patients with combined cardiac, pulmonary, and renal function abnormalities; (4) Patients with pathological conditions; and (5) Patients with incomplete pathological and clinical data. The research was endorsed by the hospital’s Ethics Committee, and all participants provided informed consent.

Reagents and instruments

Reagents: (1) PES1 antibody. Select a highly specific and sensitive PES1 monoclonal antibody for detecting PES1 expression in tumor tissues; (2) PD-L1 antibody. Select a highly specific and sensitive PD-L1 monoclonal antibody for detecting PD-L1 expression in tumor tissues; (3) Immunohistochemistry staining kit. Contains all necessary stains such as hematoxylin, eosin, etc, reagents, antibody diluent, fixative, dehydrating agent, transparency agent, etc., for immunohistochemical staining of tumor tissues; (4) PBS. Use to wash the samples and remove excess antibodies and stains; (5) Sealing agent. Use to seal the samples, protect the samples, and prevent discoloration; and (6) Pipette. Use for the precise addition of samples, such as antibodies and staining solutions.

Instruments: (1) Microscope. For observing the results of immunohistochemical staining and evaluating the expression of PES1 and PD-L1 in tumor tissues; (2) Thermostatic incubator. For temperature control during fixation, dehydration, transparency, and staining of the samples; (3) Paraffin slicer. For cutting paraffin-embedded tumor tissue blocks into thin slices; and (4) Tissue dehydrator. For the gradual transition of the tissue samples from a watery state to solvents such as alcohol and xylene to facilitate paraffin embedding.

Sample collection

Representative tumor tissues from patients with GC and HNSCC were obtained under sterile conditions by surgical resection or biopsy. Surgically resected tumor tissues were immediately placed in formalin fixative, and the fixation time was usually 24-48 h. After fixation, the tissue samples were processed into paraffin blocks through dehydration, clarification, wax immersion, and embedding procedures and stored in a 4 °C refrigerator for a long time. Before immunohistochemical testing, 4-6 μm thick slices were cut out from the paraffin blocks, attached to anti-detachment slides, and dried at 60 °C.

Immunohistochemical testing

Sections were immersed twice in xylene for 10 min each time to remove paraffin and then sequentially hydrated by passing through a gradient of 1000 g/L, 950 g/L, 900 g/L, 800 g/L, and 700 g/L ethanol for 5 min each time. Finally, the sections were washed with distilled water to remove any residual ethanol.

Antigen repair

Sections were positioned in citrate buffer (pH 6.0) and thermally repaired with antigen using a microwave oven or an autoclave to increase antigen exposure and antibody binding efficiency. After repair was completed, the sections were allowed to cool to ambient temperature and subsequently rinsed with PBS three times, each for a duration of 5 min.

Sections were treated with 30 g/L hydrogen peroxide solution and incubated at room temperature for 10 min to quench endogenous peroxidase activity, followed by rinsing with PBS three times, each for 5 min. Sections were closed using 50 g/L goat serum or bovine serum albumin and incubated at ambient temperature for a duration of 30 min to minimize nonspecific binding.

Appropriate dilution ratios of PES1 and PD-L1 specific primary antibodies were added dropwise to the sections, ensuring that the antibodies covered the tissue area evenly. The sections were placed in a wet box and incubated overnight at 4 °C or for 1-2 h at ambient temperature. The sections were washed with PBS three times, each time for 5 min to remove the unbound primary antibody.

Horseradish peroxidase-labeled secondary antibody was added dropwise on the sections and incubated at room temperature for 30 min to allow the reaction to proceed. The sections were washed with PBS 3 times for 5 min each time. Freshly prepared DAB staining solution was added by drops on the sections. The staining was observed under the microscope and immediately rinsed with distilled water to terminate the staining when a brown precipitate appeared.

Sections were re-stained using hematoxylin to color the nuclei, followed by hydrochloric acid alcohol differentiation and ammonia return to blue. Sections were subjected to graded ethanol dehydration (700 g/L, 800 g/L, 900 g/L, 950 g/L, 1000 g/L) and xylene transparency. Neutral gum was used to seal the sections, ensuring that the sections were flat and free of air bubbles. The slices were observed under a microscope, and the staining of PES1 and PD-L1 was recorded. The positive expression rate was evaluated according to the staining intensity and distribution range.

Observation indicators

(1) Expression of PES1 and PD-L1 in GC and HNSCC tissues; and (2) Association between PES1 and PD-L1 expression levels and the clinical characteristics of GC and HNSCC.

Statistical analysis

Statistical analysis was conducted using SPSS 25.0. Measurement data that followed a normal distribution were presented as mean ± SD. Comparisons between groups were made using the t test. Counting data were represented as n (%), with the χ2 test employed for analysis. The immunohistochemical staining expression of PES1 and PD-L1 proteins in GC and HNSCC tissues was evaluated using Fisher’s discriminant method (Fisher’s test). Statistical significance was set at P < 0.05.

RESULTS
Expression of PES1 and PD-L1 in GC and HNSCC tissues

Among 58 cases of GC and HNSCC, 30 cases were positive and 28 cases were negative for PES1 expression. In addition, 34 cases were positive and 24 cases were negative for PD-L1 expression. The positive expression rates of PES1 and PD-L1 were 51.72% and 58.62%, respectively (Table 1).

Table 1 Expression of pescadillo ribosomal biogenesis factor 1 and programmed death-ligand 1 in gastric cancer and head and neck squamous cell cancer tissues.

Negatives
Positive
PES128 (48.28)30 (51.72)
PD-L124 (41.38)34 (58.62)
Association between PES1 and PD-L1 expression levels and the clinical characteristics of GC and HNSCC

PES1 expression correlated with TNM stage, lymph node metastasis, and infiltration depth (P < 0.05), while it was not correlated with gender, age, degree of differentiation, tumor diameter, and diabetes mellitus (P > 0.05). PD-L1 expression was correlated with degree of differentiation, lymph node metastasis, and infiltration depth (P < 0.05) but was not correlated with gender, age, TNM stage, tumor diameter, and diabetes (P > 0.05) (Tables 2 and 3).

Table 2 Relationship between pescadillo ribosomal biogenesis factor 1 expression and clinical features of gastric cancer and head and neck squamous cell carcinoma.
Diagnostic trait
n
PES1
χ2
P value
Negative (n = 28)
Positive (n = 30)
Sex0.2220.637
    Male3717 (45.95)20 (54.05)
    Women2111 (52.38)10 (47.62)
Age (years)0.2100.647
    < 604119 (46.34)22 (53.66)
    ≥ 60179 (52.94)8 (47.06)
Degree of differentiation0.6720.412
    Poorly differentiated3217 (53.13)15 (46.88)
    Moderately to well differentiated2611 (42.31)15 (57.69)
TNM staging8.3120.004
    I-II2819 (67.86)9 (32.14)
    III309 (30.00)21 (70.00)
Lymphatic node transfer5.2870.021
    Yes3915 (38.46)24 (61.54)
    No1913 (68.42)6 (31.58)
Infiltration depth6.8460.009
    T1/T22517 (68.00)8 (32.00)
    T3/T43311 (33.33)22 (66.67)
Tumor diameter (cm)0.4850.486
    < 54121 (51.22)20 (48.78)
    ≥ 5177 (41.18)10 (58.82)
Diabetes0.0210.885
    Yes157 (46.67)8 (53.33)
    No4321 (48.84)22 (51.16)
Table 3 Relationship between programmed death-ligand 1 expression and clinical characteristics of gastric cancer and head and neck squamous cell carcinoma.
Diagnostic trait
n
PD-L1
χ2
P value
Negative (n = 24)
Positive (n = 34)
Sex0.8790.349
    Male3717 (45.95)20 (54.05)
    Women217 (33.33)14 (66.67)
Age (years)0.3670.545
    < 604118 (43.90)23 (56.10)
    ≥ 60176 (35.29)11 (64.71)
Degree of differentiation6.5080.011
    Poorly differentiated3218 (56.25)14 (43.75)
    Moderately to well differentiated266 (23.08)20 (76.92)
TNM staging0.0490.825
    I-II2812 (42.86)16 (57.14)
    III3012 (40.00)18 (60.00)
Lymphatic node transfer5.5250.019
    Yes3912 (30.77)27 (69.23)
    No1912 (63.16)7 (36.84)
Infiltration depth9.2690.002
    T1/T22516 (64.00)9 (36.00)
    T3/T4338 (24.24)25 (75.76)
Tumor diameter (cm)0.0000.984
    < 54117 (41.46)24 (58.54)
    ≥ 5177 (41.18)10 (58.82)
Diabetes1.1920.275
    Yes158 (53.33)7 (46.67)
    No4316 (37.21)27 (62.79)
DISCUSSION

GC ranks as one of the foremost causes of cancer-related fatalities on a global scale[12,13]. The majority of patients with GC are in advanced stages at the time of diagnosis and have shorter survival times and poor overall quality of life[14]. Traditional treatments include surgery, radiotherapy, and chemotherapy, but for advanced GC the cure rate of surgery is low, and the efficacy of radiotherapy is poor. Over the past few years, the development of immunotherapy and the achievement of certain clinical efficacy have made it a new treatment mode, in which immune checkpoint inhibitors play an important role[15-17].

PES1 is a nucleolin protein and has gradually been paid attention to in recent years[18]. It has been noted that in GC cell lines (AGS and N87) inhibition of PES1 expression leads to cell cycle arrest in the G1 phase, a finding that implies that low expression levels of PES1 play a part in inhibiting cell proliferation[19]. However, PES1 has not received sufficient attention within the field of GC research, and its specific significance in the clinical diagnosis and prognostic assessment of GC remains unclear.

HNSCC is a routine malignant tumor that is highly heterogeneous[20]. Although classical tumor TNM staging has been widely used for clinical diagnosis and to assist in clinical treatment decisions, TNM staging is based only on the tumor itself and fails to accurately predict the projection of patients with HNSCC. More and more studies have shown that various immune cells in the tumor microenvironment, especially B cells, contribute significantly to tumor progression and prediction of outcomes[21-24]. However, the presentation of PES1 in HNSCC and its relationship with immune infiltration have not been clarified.

In view of this, GC and HNSCC were chosen as the subjects of this study, covering two common malignant tumors, thus expanding the scope of the study. For the detection method, we used monoclonal antibodies with high specificity and sensitivity for immunohistochemical detection, which improved the accuracy of the results. In addition, this study explored the relationship between PES1 and PD-L1 for the first time, which provided new ideas for future research on the synergistic role of both in tumor immune escape.

In this study, 58 GC and HNSCC tissues were examined, and the experiments established that PES1 and PD-L1 had positive expression rates of 51.72% and 58.62%, respectively, in both tumor tissues. This finding suggests that PES1 and PD-L1 may contribute significantly to the development and progression of GC and HNSCC. Specifically, the expression level of PES1 may change with the increase of tumor malignancy and may be involved in the process of tumor invasion and metastasis. It has been shown that the high expression of PES1 in tumor tissues may be closely related to tumor development and progression, and as a potential biomarker for some tumors, it serves the purpose of early diagnosis, prognostic assessment, and monitoring of therapeutic efficacy[25-27].

PES1 is an important biomarker, and its aberrant expression has been in line with the onset and progression of a variety of cancers. In some tumors, high expression of PES1 tends to predict a more malignant, aggressive, and metastatic tumor. Therefore, PES1 may be a powerful indicator for assessing the prognosis of tumor patients. In addition, the expression of PES1 has certain specificity in the tissues of different tumors and makes its application in individualized therapy a great potential. In this study, we found that PES1 was contingent upon high expression rates in tumor tissues of both GC and HNSCC, a result that further supports the value of PES1 as a potential biomarker.

In terms of PD-L1, the findings of this research indicated a substantial association between its expression and the degree of differentiation, lymph node metastasis, and depth of infiltration of the tumor (P < 0.05). This finding is in accordance with earlier studies and suggests that PD-L1 is vital to tumor immune escape. PD-L1 inhibits T cell activation and proliferation by binding to PD-1 on the surface of T cells, which in turn helps tumor cells to dodge surveillance and clearance by the immune system[28-30]. This immune escape mechanism enables tumors to continue to grow under the suppression of the immune system and exhibits strong invasiveness and metastatic ability. Therefore, high PD-L1 expression is usually highly tied to malignant biological behaviors of tumors, such as high tumor aggressiveness, high metastatic capacity, and poor prognosis.

However, unlike PES1, PD-L1 expression showed no significant association with TNM staging (P > 0.05). This result suggests that more complex factors may need to be considered when assessing the clinical significance of PD-L1. For example, the tumor microenvironment, immune cell infiltration, and other immune escape mechanisms may have an impact on the expression level of PD-L1. Therefore, the independence of PD-L1 expression from TNM staging also suggests that relying solely on PD-L1 as a prognostic marker may have certain limitations and that multiple factors must be combined to more accurately predict patient prognosis.

In addition, this study also found that PD-L1 expression was independent of patient gender, age, tumor diameter, and diabetes mellitus status (P > 0.05), which further supported the exact function of PD-L1 in tumor immunomodulation. These results suggest that the important role played by PD-L1 in the immune escape process is not affected by these common clinical features and makes its potential and application value in tumor immunotherapy even more prominent.

Combined with the observations made in this research, we can speculate that PES1 and PD-L1 may serve as potential biomarkers for GC and HNSCC and that their high expression may be intertwined with tumor invasiveness, metastatic ability, and poor prognosis. Recent studies have shown that immune checkpoint molecules play a key role in tumor immune escape. PD-L1, as an important immune checkpoint molecule, inhibits the activation and proliferation of T cells by binding to PD-1 on the surface of T cells, which in turn helps tumor cells to evade the surveillance and clearance of the immune system.

The abnormal expression of PES1 in tumors may be related to immune regulation. In this study, we found that PES1 and PD-L1 had certain positive expression rates in GC and HNSCC tissues, and the expression of PES1 was related to TNM staging, lymph node metastasis and infiltration depth, while the expression of PD-L1 was related to differentiation degree, lymph node metastasis, and infiltration depth. This suggests that PES1 and PD-L1 may interact with each other and participate in the immune escape process of the tumor. This finding provides new ideas and directions for clinical selection of appropriate therapies, especially in the context of immunotherapy, and PES1 and PD-L1 may provide valuable references for individualized treatment. However, more large-sample studies and in-depth mechanistic studies are needed to verify this hypothesis.

Notably, although this study revealed the expression of PES1 and PD-L1 in GC and HNSCC and their relationship with clinical features, their interaction has not been directly explored yet. In view of this, future studies could further explore the interaction mechanism between PES1 and PD-L1. From the theoretical analysis, PES1 may regulate the expression level of PD-L1 by affecting the signaling pathways in tumor cells and thus regulating the expression level of PD-L1. For example, PES1 may promote the transcription of the PD-L1 gene by affecting the activity of certain transcription factors, thereby increasing the protein expression of PD-L1. In addition, PES1 may also indirectly affect the function of PD-L1 by influencing immune cell infiltration in the tumor microenvironment. For example, high expression of PES1 may attract more immune-suppressive cells to infiltrate into tumor tissues, and these immune-suppressive cells may enhance the immunosuppressive function of PD-L1 by secreting cytokines, thereby promoting immune escape from the tumor.

An in-depth study of the interaction between PES1 and PD-L1 will not only help to further reveal the mechanism of tumor immune escape but may also provide an important theoretical basis for the development of new tumor immunotherapy strategies. For example, if we can find the key targets to block the interaction between PES1 and PD-L1, it may provide new ideas for combination immunotherapy, thus improving the effectiveness of tumor treatment. In addition, exploring whether PES1 and PD-L1 can be used as biomarkers for joint prediction of tumor prognosis is also an important direction for future research. By gaining a deeper understanding of the roles of these two in tumor immunomodulation, we may be able to develop more precise and effective therapeutic strategies to further enhance the efficacy of tumor therapy and the quality of patient survival.

CONCLUSION

This study provided valuable information on the articulation of PES1 and PD-L1 in GC and HNSCC and their clinical significance. However, the present study has some limitations, such as a small sample size, which limits the generalizability and reliability of the findings. In addition, the current study only conducted univariate analysis and did not consider the effects of potential confounders such as age and gender. Therefore, future studies need to further expand the sample size and employ multifactorial regression analyses to more comprehensively reveal the roles of PES1 and PD-L1 in tumor biology to more accurately assess the relationship between their expression and the clinical features of GC and HNSCC.

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, Grade B

Novelty: Grade B, Grade C

Creativity or Innovation: Grade B, Grade C

Scientific Significance: Grade C, Grade C

P-Reviewer: Ohtsuka M; Saiura A S-Editor: Qu XL L-Editor: Filipodia P-Editor: Wang WB

References
1.  Jin M, Zhang G, Wang S, Zhao R, Zhang H. ISL1 and AQP5 complement each other to enhance gastric cancer cell stemness by regulating CD44 expression. Transl Cancer Res. 2024;13:5484-5496.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
2.  Hadebe B, Harry L, Gabela L, Masikane S, Patel M, Zwane S, Pillay V, Bipath P, Cebekhulu N, Nyakale N, Ramdass P, Msimang M, Aldous C, Sathekge M, Vorster M. Chemokine Receptor-4 Targeted PET/CT Imaging with (68)Ga-Pentixafor in Head and Neck Cancer-A Comparison with (18)F-FDG and CXCR4 Immunohistochemistry. Diagnostics (Basel). 2024;14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
3.  Deng C, Deng G, Chu H, Chen S, Chen X, Li X, He Y, Sun C, Zhang C. Construction of a hypoxia-immune-related prognostic panel based on integrated single-cell and bulk RNA sequencing analyses in gastric cancer. Front Immunol. 2023;14:1140328.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
4.  Zebene ED, Lombardi R, Pucci B, Medhin HT, Seife E, Di Gennaro E, Budillon A, Woldemichael GB. Proteomic Analysis of Biomarkers Predicting Treatment Response in Patients with Head and Neck Cancers. Int J Mol Sci. 2024;25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
5.  Nakamura K, Yaguchi T, Murata M, Ota Y, Mikoshiba A, Kiniwa Y, Okuyama R, Kawakami Y. Tumor eradication by triplet therapy with BRAF inhibitor, TLR 7 agonist, and PD-1 antibody for BRAF-mutated melanoma. Cancer Sci. 2024;115:2879-2892.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
6.  Chen H, Wei J, Zhu Z, Hou Y. Multifaceted roles of PD-1 in tumorigenesis: From immune checkpoint to tumor cell-intrinsic function. Mol Carcinog. 2024;63:1436-1448.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
7.  Oualla K, Castelo Branco L, Nouiyakh L, Amaadour L, Benbrahim Z, Arifi S, Mellas N. Therapeutic Approaches With Immune Checkpoint Inhibitors in Head and Neck Cancers and the Role of PD-L1 as a Biomarker. Cancer Control. 2021;28:10732748211004878.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
8.  Elbehi AM, Anu RI, Ekine-Afolabi B, Cash E. Emerging role of immune checkpoint inhibitors and predictive biomarkers in head and neck cancers. Oral Oncol. 2020;109:104977.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11]  [Cited by in RCA: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
9.  Novoplansky O, Jagadeeshan S, Prasad M, Yegodayev KM, Marripati D, Shareb RA, Greenshpan Y, Mathukkada S, Ben-Lulu T, Bhattacharya B, Porgador A, Kong D, Brägelmann J, Gutkind JS, Elkabets M. Dual inhibition of HERs and PD-1 counteract resistance in KRAS(G12C)-mutant head and neck cancer. J Exp Clin Cancer Res. 2024;43:308.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
10.  Laban S, Remark R, Idel C, Ribbat-idel J, Krupar R, Schröck A, Klümper N, Döscher J, Sikora A, Abou Kors T, von Witzleben A, Vahl J, Grages A, Sonntag M, Brunner C, Hoffmann T, Gnjatic S. 915P Longer OS and RFS for CD3high/PD-L1+ head and neck squamous cell carcinoma (HNSCC) patients. Ann Oncol. 2024;35:S643.  [PubMed]  [DOI]  [Full Text]
11.  Yuan S, Xu N, Yang J, Yuan B. Emerging role of PES1 in disease: A promising therapeutic target? Gene. 2025;932:148896.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
12.  Li XQ, Yang J, Liu B, Han SM. Disitamab vedotin combined with apatinib in gastric cancer: A case report and review of literature. World J Clin Oncol. 2024;15:1351-1358.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
13.  Liu Y, Yu X, Shen H, Hong Y, Hu G, Niu W, Ge J, Xuan J, Qin JJ, Li Q. Mechanisms of traditional Chinese medicine in the treatment and prevention of gastric cancer. Phytomedicine. 2024;135:156003.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
14.  Tang L, He Y, Pang Y, Su Z, Zhou Y, Wang Y, Lu Y, Jiang Y, Han X, Song L, Wang L, Li Z, Lv X, Wang Y, Yao J, Liu X, Zhou X, He S, Zhang Y, Song L, Li J, Wang B. Suicidal ideation in advanced cancer patients without major depressive disorder. Psychooncology. 2022;31:1941-1950.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
15.  Yang T, Xu R, You J, Li F, Yan B, Cheng JN. Prognostic and clinical significance of HER-2 low expression in early-stage gastric cancer. BMC Cancer. 2022;22:1168.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Reference Citation Analysis (0)]
16.  Araruna GF, Ribeiro HSC, Torres SM, Diniz AL, Godoy AL, Farias IC, Costa WL Jr, Coimbra FJF. Impact of Minimally Invasive Surgery on Early and Late Outcomes of Patients With Gastric Cancer Treated Using Neoadjuvant Chemotherapy. J Surg Oncol. 2024;130:776-784.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
17.  Zhou Y, Wang Q, Deng H, Xu B, Zhou Y, Liu J, Liu Y, Shi Y, Zheng X, Jiang J. N6-methyladenosine demethylase FTO promotes growth and metastasis of gastric cancer via m(6)A modification of caveolin-1 and metabolic regulation of mitochondrial dynamics. Cell Death Dis. 2022;13:72.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 41]  [Cited by in RCA: 45]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
18.  Li YZ, Zhang C, Pei JP, Zhang WC, Zhang CD, Dai DQ. The functional role of Pescadillo ribosomal biogenesis factor 1 in cancer. J Cancer. 2022;13:268-277.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
19.  Li J, Zhou X, Lan X, Zeng G, Jiang X, Huang Z. Repression of PES1 expression inhibits growth of gastric cancer. Tumour Biol. 2016;37:3043-3049.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 7]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
20.  Bhat AA, Yousuf P, Wani NA, Rizwan A, Chauhan SS, Siddiqi MA, Bedognetti D, El-Rifai W, Frenneaux MP, Batra SK, Haris M, Macha MA. Tumor microenvironment: an evil nexus promoting aggressive head and neck squamous cell carcinoma and avenue for targeted therapy. Signal Transduct Target Ther. 2021;6:12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 25]  [Cited by in RCA: 91]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
21.  Toh J, Reitsma AJ, Tajima T, Younes SF, Ezeiruaku C, Jenkins KC, Peña JK, Zhao S, Wang X, Lee EYZ, Glass MC, Kalesinskas L, Ganesan A, Liang I, Pai JA, Harden JT, Vallania F, Vizcarra EA, Bhagat G, Craig FE, Swerdlow SH, Morscio J, Dierickx D, Tousseyn T, Satpathy AT, Krams SM, Natkunam Y, Khatri P, Martinez OM. Multi-modal analysis reveals tumor and immune features distinguishing EBV-positive and EBV-negative post-transplant lymphoproliferative disorders. Cell Rep Med. 2024;5:101851.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
22.  Wang B, Liu W, Song B, Li Y, Wang Y, Tan B. Targeting LINC00665/miR-199b-5p/SERPINE1 axis to inhibit trastuzumab resistance and tumorigenesis of gastric cancer via PI3K/AKt pathway. Noncoding RNA Res. 2025;10:153-162.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
23.  Mi D, Li C, Li Y, Yao M, Li Y, Hong K, Xie C, Chen Y. Discovery of novel BCL6-Targeting PROTACs with effective antitumor activities against DLBCL in vitro and in vivo. Eur J Med Chem. 2024;277:116789.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
24.  Low JT, Christie M, Ernst M, Dumoutier L, Preaudet A, Ni Y, Griffin MDW, Mielke LA, Strasser A, Putoczki TL, O'Reilly LA. Loss of NFKB1 Results in Expression of Tumor Necrosis Factor and Activation of Signal Transducer and Activator of Transcription 1 to Promote Gastric Tumorigenesis in Mice. Gastroenterology. 2020;159:1444-1458.e15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 18]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
25.  Yao X, Lu C, Shen J, Jiang W, Qiu Y, Zeng Y, Li L. A novel nine gene signature integrates stemness characteristics associated with prognosis in hepatocellular carcinoma. BIOCELL. 2021;45:1425-1448.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
26.  He Y, Xiang J, Li Y, Huang W, Gu F, Wang Y, Chen R. PES1 is a biomarker of head and neck squamous cell carcinoma and is associated with the tumor microenvironment. Cancer Med. 2023;12:12622-12638.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
27.  Fu Z, Jiao Y, Li YQ, Ke JJ, Xu YH, Jia BX, Liu B. PES1 In Liver Cancer: A Prognostic Biomarker With Tumorigenic Roles. Cancer Manag Res. 2019;11:9641-9653.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
28.  Albrecht T, Brinkmann F, Albrecht M, Lonsdorf AS, Mehrabi A, Hoffmann K, Kulu Y, Charbel A, Vogel MN, Rupp C, Köhler B, Springfeld C, Schirmacher P, Roessler S, Goeppert B. Programmed Death Ligand-1 (PD-L1) Is an Independent Negative Prognosticator in Western-World Gallbladder Cancer. Cancers (Basel). 2021;13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 16]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
29.  Fokter Dovnik N, Smyth EC. Changes in the therapeutic landscape of oesophago-gastric cancers. Curr Opin Oncol. 2021;33:362-367.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
30.  Bansal A, Srinivasan R, Rohilla M, Rai B, Rajwanshi A, Suri V, Chandra Saha S. Immunotyping in tubo-ovarian high-grade serous carcinoma by PD-L1 and CD8+ T-lymphocytes predicts disease-free survival. APMIS. 2021;129:254-264.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]