Open Access

Clinical prognostic significance of xeroderma pigmentosum group C and IFN‑γ in non‑small cell lung cancer

  • Authors:
    • Yongming Wang
    • Weiyu Wang
    • Huaijie Wang
    • Liya Qin
    • Meijia Zhang
    • Yong Zhang
    • Yubing Wang
    • Changcheng Hao
    • Meihua Qu
    • Gongchao Wang
  • View Affiliations

  • Published online on: April 9, 2024     https://doi.org/10.3892/ol.2024.14392
  • Article Number: 259
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer is the most common cancer in the world due to its high incidence and recurrence. Genetic instability is one of the main factors leading to its occurrence, development and poor prognosis. Decreased xeroderma pigmentosum group C (XPC) expression notably enhances the stem cell properties of lung cancer cells and increases their proliferation and migration. Additionally, patients with lung cancer and low XPC expression had a poor prognosis. The purpose of the present study was to analyze the effect of XPC and IFN‑γ on the clinical prognosis of patients with non‑small cell lung cancer (NSCLC). Lung adenocarcinoma specimens were collected from a total of 140 patients with NSCLC. Additionally, from these 140 patients, 48 paracarcinoma tissue specimens were also collected, which were later used to construct tissue microarrays. The expression of XPC and IFN‑γ in cancer tissues and in paraneoplastic tissues was detected using immunohistochemistry. The prognosis and overall survival of patients were determined through telephone follow‑up. The results showed a positive correlation between expression of XPC and IFN‑γ in NSCLC. Additionally, high expression of both markers was associated with a favorable prognosis in patients with NSCLC. The aforementioned findings suggest that the expression of XPC and IFN‑γ has prognostic value in clinical practice and is expected to become a marker for clinical application.

Introduction

Lung cancer causes 25% of cancer-related deaths and has a high incidence rate despite significant advances in therapies made in recent years, representing 11.6% of new cancer cases worldwide (1,2). Genomic instability is one of the main factors leading to tumor occurrence, progression and poor outcomes (2). Xeroderma pigmentosum group C (XPC) is a DNA damage recognition factor and a promoter of nucleotide excision repair that plays an important role in maintaining DNA stability (3). XPC is a key factor in the development of a number of tumor types including lung cancer (47). Our previous study revealed that a reduction in XPC expression can markedly promote the stem cell properties of lung cancer cells and increase the proliferation and migration of lung cancer cells, and a low XPC expression in patients with lung cancer is associated with poor prognosis (8). Zhang et al (9) showed that the loss of XPC could increase the amount of oxidatively damaged DNA and promote Kras-mediated lung cancer development. The underlying mechanisms through which XPC affects lung cancer development remain not understood; more specifically, the mediator responsible for these processes has yet to be identified.

IFN-γ in the blood comes from monocytes, macrophages, endothelial cells and tumor cells, and considered a diagnostic and prognostic biomarker in lung cancer (10,11). Studies have shown that IFN-γ can mediate lung cancer inhibition via programmed death-ligand 1 expression (12,13). IFN-γ-induced endoplasmic reticulum stress impairs autophagy and triggers apoptosis in lung cancer cells (14).

A total of >90% of lung cancer-related deaths are due to metastasis of lung cancer cells (15). Cytological studies have shown that the expression of both XPC and IFN-γ is closely related to the metastasis of lung cancer (8,16).

A study by Wu et al (17) revealed that XPC expression levels were linked to prognosis and metastasis in patients with lung cancer. In a study by Ahn et al (18) investigating the relationship between the expression level of IFN-γ, and the prognosis and metastasis status of patients with adenocarcinoma, it was found that patients with low in vitro production of IFN-γ by peripheral immune cells had a markedly lower 1-year overall survival rate than patients with high IFN-γ production.

However, the expression of XPC and IFN-γ in lung cancer tissue has yet to be studied. In the present study, 140 lung cancer tissue samples were collected, and the correlation between the expression of XPC and IFN-γ in lung cancer tissue and patient prognosis was analysed. In addition, the differences in the expression of XPC and IFN-γ were investigated according to the histopathological characteristics of different patients with lung cancer.

Materials and methods

The Cancer Genome Atlas (TCGA) analysis

Using the relevant clinical data of TCGA database, the relationship between the expression of XPC and the clinicopathological characteristics of patients with lung cancer was statistically analysed. There are no data on IFN-γ in TCGA database. The present study complied with the National Institutes of Health TCGA Human Guidelines Subject Protection and Data Access Policy, and the results shown are in whole or part based upon data generated by the TCGA Research Network: cancer.gov/tcga (19).

Specimens

Wax blocks of lung adenocarcinoma tissues from patients with lung cancer with a pathological diagnosis were collected from Weifang Second People's Hospital (Weifang, China) between January 2011 and December 2015. Confirmation of diagnosis was conducted by two independent pathologists using H&E staining. For this, tissues were fixed using 4% paraformaldehyde at 25°C for 24 h and embedded in paraffin. Sections were cut into 5-µm thick slices and mounted on adhesive slides using a sectioning machine. The slices were then baked at 60°C for 2 h. Subsequently, the tissue sections were rehydrated using xylene and a gradient of 100-70% alcohol, stained with hematoxylin at room temperature for 7 min and eosin at room temperature for 1 min. Finally, the sections were dehydrated in an increasing series of alcohol (70–100%) and sealed with a neutral resin. Observations were conducted using a light microscope under a 20× objective and 10× eyepiece. Tumor specimens were collected from 140 patients, from which 48 paracancerous samples were also obtained, and made into one tissue microchip by Shanghai Outdo Biotech Co., Ltd. The tissue microchip was used for subsequent experiments. Patient prognosis and survival time were obtained via telephone follow-up.

The present study protocols were reviewed and approved by the ethics committee of Weifang Second People's Hospital (YX2020-001-01). The study complied with the relevant regulations of the Declaration of Helsinki and was conducted under the supervision of the hospital ethics committee.

Tissue microarray and immunohistochemistry (IHC)

Immunohistochemical staining with XPC and IFN-γ antibodies was performed on a microchip to analyse the protein expression in NSCLC tissues. The tissues embedded in paraffin, which were used for HE staining in the previous step, were processed into a tissue chip using a tissue microarray spotter. The tissue chip was then baked at 55°C for 4 h to fix the tissue. Subsequently, the tissue chip was sliced into 4-µm wax slices using a tissue slicer and affixed to adhesive slides. After xylene and gradient alcohol treatment to rehydrate the tissues, the tissues were repaired by microwave heating and boiling in Citrate Antigen Repair Solution (3.23 g/l; pH, 6; cat. no. ZLI-9064; Beijing Zhongshan Jinqiao Biotechnology Co.) for 30 min, followed by incubation with 3% hydrogen peroxide for 5 min at room temperature, and then incubation with 3% goat serum (cat. no. SL038; Sorabio) for 1 h. Subsequently, the tissues were incubated with either XPC Rabbit Polyclonal Primary Antibody (1:50; cat. no. ab155025; Abcam) or IFN-γ Rabbit Polyclonal Primary Antibody (1:1,000; cat. no. ab25101; Abcam) overnight (>12 h) at 4°C. The tissue sections were then incubated with an HRP-conjugated goat-conjugated anti-rabbit secondary antibody (1:500; cat. no. ab6721; Abcam) for 4 h at room temperature. Hematoxylin staining (cat. no. H8070; Sorabio) at room temperature for 7 min. Finally, sections were dehydrated in an increasing series of alcohol (70–100%) and sealed with a neutral resin. The IHC images were collected using a Panoramic MIDI light microscope (3DHISTECH, Ltd.), and the percentage of positive IHC signals was analyzed using Density Quant version 2.3 (3DHISTECH, Ltd.). According to the IHC staining intensity, samples were grouped according to the percentage of positive cells. A positive rate >90% was considered to indicate high expression and was marked as XPChigh or IFN-γhigh; a positive rate <90% was considered to indicate low expression and was marked as XPClow or IFN-γlow.

Statistical analysis

SPSS (version 22; IBM Corp.) was used to perform a χ2-test and analyze the expression of XPC and IFN-γ in NSCLC tissues, and the relationship between the expression of XPC or IFN-γ and the clinicopathological characteristics of patients with NSCLC. The association between XPC and IFN-γ was calculated using Pearson's correlation coefficient. A Kaplan-Meier survival curve was used to perform survival analysis, and the log-rank test was applied to evaluate statistical significance. A unpaired t-test was used to compare two groups of TCGA data, while one-way ANOVA was used to analyze the TCGA data from ≥3 groups. The log-rank test was used for survival curve analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

XPC is expressed at low levels in lung adenocarcinoma tissues in TCGA database

The expression of XPC, a nucleic acid splice repair gene, was significantly different in various cancers (Fig. 1A). In the present study, the relationship between the expression of XPC in 502 lung cancer tissues in TCGA database and patient prognosis was first analyzed. The results showed that patients with high XPC expression had a better prognosis than those patients with low XPC expression; however, the difference was not significant (P=0.083; Fig. 1B).

In addition, the expression of XPC in 574 lung cancer tissues including 515 lung cancer tissues and 59 adjacent normal tissues was analysed. Box plots revealed that the expression of XPC in lung tumor tissues was significantly different; compared with the XPC expression in normal tissues, XPC expression was significantly lower in lung tumor tissues (Fig. 1C). In addition, the expression of XPC in lung tumor tissues was not related to sex (Fig. 1D).

Expression of XPC and IFN-γ is positively correlated in the tumor tissues of patients with NSCLC

Immunohistochemical images of samples from all patients are presented in Fig. 2. The area encircled by the black box represents a sample of paracancerous tissue. Based on the calculation method of Pearson's correlation coefficient mentioned by Akoglu H (20), after Pearson's correlation coefficient, the results showed a correlation between XPC and IFN-γ in the tissues of patients with NSCLC. To investigate whether the expression of XPC was associated with that of IFN-γ in the tissues of patients with NSCLC, immunohistochemical analysis using cancer tissues from 140 patients with NSCLC was performed. There was a significant positive correlation between the expression of XPC and that of IFN-γ. In case 1, XPC and IFN-γ were expressed at high levels (Fig. 3A). In case 2, both XPC and IFN-γ were moderately expressed (Fig. 3B). In case 3, the expression levels of XPC and IFN-γ were weak (Fig. 3C). The expression of XPC and IFN-γ in NSCLC tissues was significantly positively correlated (r=0.395; P<0.01; Fig. 3D).

XPC and IFN-γ expression is associated with a better prognosis in patients with NSCLC

Given the correlation between the expression of XPC and IFN-γ in NSCLC tissues, the relationship between the co-expression of XPC and IFN-γ and the prognosis of patients with NSCLC was analyzed. Our previous study revealed that high XPC expression was positively correlated with a good prognosis in patients (8). High expression of IFN-γ was positively correlated with good patient prognosis (log-rank; P=0.0368, Fig. 4A), whereas analysis of XPC expression alone also showed a significant positive correlation between XPC expression and good patient prognosis (log-rank; P=0.0419, Fig. 4B). In addition, based on the expression of XPC and IFN-γ, patients were divided into the XPChighIFN-γhigh and XPClowIFN-γlow groups and Kaplan-Meier survival analysis was carried out. The survival time of patients in the XPChighIFN-γhigh group was significantly longer than that of patients in the XPClowIFN-γlow group (log-rank, P=0.0326; Fig. 4C).

IFN-γ expression is associated with tumor metastasis in patients with NSCLC

To further study the significance of XPC and IFN-γ expression in the occurrence, metastasis and recurrence of NSCLC, the association of XPC and IFN-γ expression with the TNM stage was investigated (21). There was no significant difference in the expression of IFN-γ at different tumor stages or N-phases (Fig. 5), but there was a significant difference in the expression of IFN-γ with tumor metastasis (P=0.036; Fig. 5H). However, XPC expression was not strongly associated with patient TNM stage.

Discussion

Lung cancer can be divided into SCLC and NSCLC according to histological classification (22). NSCLC is the most common subtype of lung cancer and can be divided into adenocarcinoma, squamous cell cancer and large-cell lung cancer. With its high incidence and recurrence, a better understanding of NSCLC is required for lung cancer treatment (2326).

Genomic instability is the primary factor promoting the occurrence and malignant transformation of cancer (27). XPC is a protein that recognizes damage and is involved in the nucleotide excision repair pathway (7,28,29). Studies have shown that low XPC expression is associated with poor outcomes in patients with NSCLC (8,30). However, a study by Teng et al (31) showed that upregulation of XPC expression could increase the resistance of lung cancer cells to the chemotherapeutic drug cisplatin through the PI3K/AKT signaling pathway. This phenomenon has not been validated in animal models or clinical studies. It is hypothesized that the complex tumor microenvironment and immune microenvironment in tumor tissues can affect the physiological function of tumors. Cytological and animal experiments have shown that reduced XPC expression can promote lung cancer cell proliferation and migration (32).

IFN-γ is a key moderator of cellular immunity and is secreted by activated T lymphocytes, gd cells and natural killer cells (33,34). Studies have indicated that IFN-γ acts as an antitumor factor by activating the immune system and triggering tumor cell apoptosis (3537). It has also been shown that IFN-γ protects tumor cells during immune checkpoint blockade (ICB) therapy by inducing CD4+ T cell apoptosis and facilitating tumor cell evasion from CD8+ T cell cytotoxicity (38). It is noteworthy that IFN-γ exerts this function as a modulatory effect during the application of ICB. In general, the biological effects of IFN receptor signaling are regulated by three main factors: i) Expression profile of IFN itself; ii) profile of the receptor; and iii) expression of the target gene (39). Activated IFNs can directly initiate gene transcription and multiple downstream signaling pathways, leading to a variety of cellular responses, such as cell cycle arrest and apoptosis in tumor cells (40). Moreover, the prognostic and pathological characteristics of patients with lung cancer need to be studied further to determine the clinical significance of IFN-γ.

Both XPC and IFN-γ play important roles in cancer progression. However, the correlation between XPC and IFN-γ has not been studied. Our previous study revealed that low XPC expression could not only promote the proliferation and migration of lung cancer cells but also increase the stem cell characteristics of lung cancer cells (8). Song et al (41) reported that low expression of IFN-γ could confer stem cell properties to lung cancer cells through the ICAM1-PI3K-AKT-NOTCH1 pathway, while high expression of IFN-γ could induce apoptosis in NSCLC through activation of the JAK1-STAT1-caspase pathway. Low levels of XPC and IFN-γ can promote the characteristics of lung cancer stem cells. To further investigate the correlation between XPC and IFN-γ expression in lung cancer tissues in the present study, the expression of XPC and IFN-γ was investigated in the tumor tissues of 140 patients with NSCLC; it was found that the expression of XPC and IFN-γ in NSCLC was significantly positively correlated. Moreover, patients with high IFN-γ expression had a favorable prognosis. The TNM staging results indicated a decreased proportion of patients with low IFN-γ expression at stage M0, suggesting distant metastasis of tumor cells. As previously discussed, both XPC and IFN-γ can significantly influence the stemness of lung cancer cells during tumor progression. Through TNM staging analysis, it was discovered that patients with high XPC and INF-γ expression were more likely to develop distal metastases. It is hypothesized that this difference may be linked to the increased stemness of the tumor cells mentioned earlier. The significant correlation between changes in the expression of the two genes suggested a potential link between them in this process. Although the survival curves indicated that the expression of both IFN-γ and XPC significantly affects patient prognosis, the sample size limited further investigations of the correlation between the two. This limitation led to the hypothesis that a more pronounced synergistic effect was not observed. Upon analyzing the data from public databases, significant variability in XPC expression in patients was observed, with standard errors >20% in some cases. While this variability is attributed to individual patient differences, it could still potentially influence patient prognosis. In future studies, the number of clinical samples will be increased and the association between the JAK/STAT pathway, and IFN-γ and tumor cell stemness will be investigated to elucidate the combined impact of the two on tumor cell stemness. These findings could lead to future studies investigating the interaction between XPC and IFN-γ and the mechanism through which XPC and IFN-γ affect the occurrence and recurrence of lung cancer.

In conclusion, the current study is the first to report the prognostic significance of the combination of XPC and IFN-γ detected by IHC in patients with lung cancer. The present study provides novel evidence that links XPC and IFN-γ expression in lung cancer. Evaluation of the expression of XPC and IFN-γ may lead to the identification of novel prognostic factors and therapeutic strategies for lung cancer.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81871892), the Science and Technology Development Project of Weifang (grant nos. 2021YX070 and 2022ZJ1059), the Science and Technology Plan of Shandong Health Committee (grant no. 2019 WSA07011) and the Scientific Project of Weifang Health Commission (grant no. WFWSJK-2022-220).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

YoW and WW were responsible for collecting and preparing pathological specimens and conducting the IHC staining. HW, LQ and MZ conducted patient visits and compiled data. YZ, YuW and CH designed the experiments and provided guidance for the writing of the manuscript. MQ and GW developed the experimental protocol, funded the study and reviewed the reliability of the data. MQ and GW confirm the authenticity of all the raw data. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

The protocols for the present study were reviewed and approved by The Ethics Committee of Weifang Second People's Hospital (Weifang, China; YX2020-001-01); all patients provided written informed consent. All analyses were conducted in accordance with the Declaration of Helsinki.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wu F, Wang L and Zhou C: Lung cancer in China: Current and prospect. Curr Opin Oncol. 33:40–46. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Bade BC and Dela Cruz CS: Lung Cancer 2020: Epidemiology, etiology, and prevention. Clin Chest Med. 41:1–24. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Shell SM, Hawkins EK, Tsai MS, Hlaing AS, Rizzo CJ and Chazin WJ: Xeroderma pigmentosum complementation group C protein (XPC) serves as a general sensor of damaged DNA. DNA Repair (Amst). 12:947–953. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Zhou H, Saliba J, Sandusky GE and Sears CR: XPC protects against smoking- and carcinogen-induced lung adenocarcinoma. Carcinogenesis. 40:403–411. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Kobaisi F, Sulpice E, Barette C, Fayyad N, Fauvarque MO, Badran B, Fayyad-Kazan M, Fayyad-Kazan H, Gidrol X and Rachidi W: Isoconazole and clemizole hydrochloride partially reverse the xeroderma pigmentosum C phenotype. Int J Mol Sci. 22:81562021. View Article : Google Scholar : PubMed/NCBI

6 

D'Errico M, Parlanti E, Teson M, de Jesus BM, Degan P, Calcagnile A, Jaruga P, Bjørås M, Crescenzi M, Pedrini AM, et al: New functions of XPC in the protection of human skin cells from oxidative damage. EMBO J. 25:4305–4315. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Fayyad N, Kobaisi F, Beal D, Mahfouf W, Ged C, Morice-Picard F, Fayyad-Kazan M, Fayyad-Kazan H, Badran B, Rezvani HR and Rachidi W: Xeroderma pigmentosum C (XPC) mutations in primary fibroblasts impair base excision repair pathway and increase oxidative DNA damage. Front Genet. 11:5616872020. View Article : Google Scholar : PubMed/NCBI

8 

Wang W, Ma S, Ding Z, Yang Y, Wang H, Yang K, Cai X, Li H, Gao Z and Qu M: XPC protein improves lung adenocarcinoma prognosis by inhibiting lung cancer cell stemness. Front Pharmacol. 12:7079402021. View Article : Google Scholar : PubMed/NCBI

9 

Zhang X, He N, Gu D, Wickliffe J, Salazar J, Boldogh I and Xie J: Genetic Evidence for XPC-KRAS interactions during lung cancer development. J Genet Genomics. 42:589–596. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Abolfathi H, Sheikhpour M, Shahraeini SS, Khatami S and Nojoumi SA: Studies in lung cancer cytokine proteomics: A review. Expert Rev Proteomics. 18:49–64. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Marrugal A, Ojeda L, Paz-Ares L, Molina-Pinelo S and Ferrer I: Proteomic-Based approaches for the study of cytokines in lung cancer. Dis Markers. 2016:21386272016. View Article : Google Scholar : PubMed/NCBI

12 

Gao Y, Yang J, Cai Y, Fu S, Zhang N, Fu X and Li L: IFN-ү-mediated inhibition of lung cancer correlates with PD-L1 expression and is regulated by PI3K-AKT signaling. Int J Cancer. 143:931–943. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Zhang X, Zeng Y, Qu Q, Zhu J, Liu Z, Ning W, Zeng H, Zhang N, Du W, Chen C and Huang JA: PD-L1 induced by IFN-ү from tumor-associated macrophages via the JAK/STAT3 and PI3K/AKT signaling pathways promoted progression of lung cancer. Int J Clin Oncol. 22:1026–1033. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Fang C, Weng T, Hu S, Yuan Z, Xiong H, Huang B, Cai Y, Li L and Fu X: IFN-ү-induced ER stress impairs autophagy and triggers apoptosis in lung cancer cells. Oncoimmunology. 10:19625912021. View Article : Google Scholar : PubMed/NCBI

15 

Nasim F, Sabath BF and Eapen GA: Lung cancer. Med Clin North Am. 103:463–473. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Kgokolo MCM, Anderson K, Siwele SC, Steel HC, Kwofie LLI, Sathekge MM, Meyer PWA, Rapoport BL and Anderson R: Elevated levels of soluble CTLA-4, PD-1, PD-L1, LAG-3 and TIM-3 and systemic inflammatory stress as potential contributors to immune suppression and generalized tumorigenesis in a cohort of South African xeroderma pigmentosum patients. Front Oncol. 12:8197902022. View Article : Google Scholar : PubMed/NCBI

17 

Wu YH, Wu TC, Liao JW, Yeh KT, Chen CY and Lee H: p53 dysfunction by xeroderma pigmentosum group c defects enhance lung adenocarcinoma metastasis via increased Mmp1 expression. Cancer Res. 70:10422–10432. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Ahn SS, Kwon M, Sung M, Jung SM, Lee SW, Park YB, Kim ST and Song JJ: Ex Vivo Interferon gamma production by peripheral immune cells predicts survival in lung adenocarcinoma. Clin Lung Cancer. 20:e299–e308. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Carrot-Zhang J, Yao X, Devarakonda S, Deshpande A, Damrauer JS, Silva TC, Wong CK, Choi HY, Felau I, Robertson AG, et al: Whole-genome characterization of lung adenocarcinomas lacking alterations in the RTK/RAS/RAF pathway. Cell Rep. 34:1087842021. View Article : Google Scholar : PubMed/NCBI

20 

Akoglu H: User's guide to correlation coefficients. Turk J Emerg Med. 18:91–93. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Asamura H, Nishimura KK, Giroux DJ, Chansky K, Hoering A, Rusch V and Rami-Porta R; Members of the IASLC Staging and Prognostic Factors Committee of the Advisory Boards, and Participating Institutions, : IASLC lung cancer staging project: The new database to inform revisions in the Ninth Edition of the TNM classification of lung cancer. J Thorac Oncol. 18:564–575. 2023. View Article : Google Scholar : PubMed/NCBI

22 

Villalobos P and Wistuba II: Lung cancer biomarkers. Hematol Oncol Clin North Am. 31:13–29. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Gao C, Kong N, Zhang F, Zhou L, Xu M and Wu L: Development and validation of the potential biomarkers based on m6A-related lncRNAs for the predictions of overall survival in the lung adenocarcinoma and differential analysis with cuproptosis. BMC Bioinformatics. 23:3272022. View Article : Google Scholar : PubMed/NCBI

24 

Ruiz-Cordero R and Devine WP: Targeted therapy and checkpoint immunotherapy in lung cancer. Surg Pathol Clin. 13:17–33. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Rodriguez-Canales J, Parra-Cuentas E and Wistuba II: Diagnosis and molecular classification of lung cancer. Cancer Treat Res. 170:25–46. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Luo C, Lei M and Zhang Y, Zhang Q, Li L, Lian J, Liu S, Wang L, Pi G and Zhang Y: Systematic construction and validation of an immune prognostic model for lung adenocarcinoma. J Cell Mol Med. 24:1233–1244. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Lundin A and Driscoll B: Lung cancer stem cells: Progress and prospects. Cancer Lett. 338:89–93. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Sugasawa K: XPC: Its Product and Biological Roles. Molecular Mechanisms of Xeroderma Pigmentosum. Advances in Experimental Medicine and Biology. Ahmad SI and Hanaoka F: Volume 637. Springer; New York, NY: 2008, PubMed/NCBI

29 

Nemzow L, Lubin A, Zhang L and Gong F: XPC: Going where no DNA damage sensor has gone before. DNA Repair (Amst). 36:19–27. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Cui T, Srivastava AK, Han C, Yang L, Zhao R, Zou N, Qu M, Duan W, Zhang X and Wang Q-E: XPC inhibits NSCLC cell proliferation and migration by enhancing E-Cadherin expression. Oncotarget. 6:10060–10072. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Teng X, Fan XF, Li Q, Liu S, Wu DY, Wang SY, Shi Y and Dong M: XPC inhibition rescues cisplatin resistance via the Akt/mTOR signaling pathway in A549/DDP lung adenocarcinoma cells. Oncol Rep. 41:1875–1882. 2019.PubMed/NCBI

32 

Nasrallah NA, Wiese BM and Sears CR: Xeroderma pigmentosum complementation group C (XPC): Emerging roles in non-dermatologic malignancies. Front Oncol. 12:8469652022. View Article : Google Scholar : PubMed/NCBI

33 

Medler TR, Murugan D, Horton W, Kumar S, Cotechini T, Forsyth AM, Leyshock P, Leitenberger JJ, Kulesz-Martin M, Margolin AA, et al: Complement C5a fosters squamous carcinogenesis and limits T cell response to chemotherapy. Cancer Cell. 34:561–578.e6. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Weichselbaum RR, Liang H, Deng L and Fu YX: Radiotherapy and immunotherapy: A beneficial liaison? Nat Rev Clin Oncol. 14:365–379. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Burke F, East N, Upton C, Patel K and Balkwill FR: Interferon gamma induces cell cycle arrest and apoptosis in a model of ovarian cancer: Enhancement of effect by batimastat. EJC. 33:1114–1121. 1997. View Article : Google Scholar : PubMed/NCBI

36 

Schoenborn JR and Wilson CB: Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol. 96:41–101. 2007.PubMed/NCBI

37 

Shin EC, Ahn JM, Kim CH, Choi Y, Ahn YS, Kim H, Kim SJ and Park JH: IFN-gamma induces cell death in human hepatoma cells through a TRAIL/death receptor-mediated apoptotic pathway. Int J Cancer. 93:262–268. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Minn AJ and Wherry EJ: Combination cancer therapies with immune checkpoint blockade: Convergence on interferon signaling. Cell. 165:272–275. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Galani V, Kastamoulas M, Varouktsi A, Lampri E, Mitselou A and Arvanitis DL: IFNs-signaling effects on lung cancer: An up-to-date pathways-specific review. Clin Exp Med. 17:281–289. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Lee KS, Chung WY, Park JE, Jung YJ, Park JH, Sheen SS and Park KJ: Interferon-ү-Inducible chemokines as prognostic markers for lung cancer. Int J Environ Res Public Health. 18:93452021. View Article : Google Scholar : PubMed/NCBI

41 

Song M, Ping Y, Zhang K, Yang L, Li F, Zhang C, Cheng S, Yue D, Maimela NR, Qu J, et al: Low-Dose IFNү induces tumor cell stemness in tumor microenvironment of non-small cell lung cancer. Cancer Res. 79:3737–3748. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2024
Volume 27 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Wang W, Wang H, Qin L, Zhang M, Zhang Y, Wang Y, Hao C, Qu M, Wang G, Wang G, et al: Clinical prognostic significance of xeroderma pigmentosum group C and IFN‑&gamma; in non‑small cell lung cancer. Oncol Lett 27: 259, 2024
APA
Wang, Y., Wang, W., Wang, H., Qin, L., Zhang, M., Zhang, Y. ... Wang, G. (2024). Clinical prognostic significance of xeroderma pigmentosum group C and IFN‑&gamma; in non‑small cell lung cancer. Oncology Letters, 27, 259. https://doi.org/10.3892/ol.2024.14392
MLA
Wang, Y., Wang, W., Wang, H., Qin, L., Zhang, M., Zhang, Y., Wang, Y., Hao, C., Qu, M., Wang, G."Clinical prognostic significance of xeroderma pigmentosum group C and IFN‑&gamma; in non‑small cell lung cancer". Oncology Letters 27.6 (2024): 259.
Chicago
Wang, Y., Wang, W., Wang, H., Qin, L., Zhang, M., Zhang, Y., Wang, Y., Hao, C., Qu, M., Wang, G."Clinical prognostic significance of xeroderma pigmentosum group C and IFN‑&gamma; in non‑small cell lung cancer". Oncology Letters 27, no. 6 (2024): 259. https://doi.org/10.3892/ol.2024.14392