This work is licensed under http://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
1. Introduction
Lung cancer is a common malignant tumor in the world. According to the latest data released by the international agency for research on cancer in CA:A Cancer Journal for Clinicians [1], there were 2.207 million new lung cancer cases and 1.796 million reported deaths worldwide, accounting for 11.7% and 18.0% of the global cancer incidence rate and mortality, respectively. Nonsmall cell lung cancer (NSCLC) has decreased in incidence overall in the past decade in the United States but the incidence of stage I NSCLC has increased along with its prevalence [2], highlighting the need for research in this area. Surgical resection can be the first choice for the treatment of NSCLC. However, more than 70% of patients with NSCLC were often accompanied by tumor proliferation and metastasis and lost the best opportunity for surgery due to the limitations of early diagnosis [3]. According to a global cancer detection report, lung cancer patients in most countries had a 5-year survival rate of only 10-20% [4]. With the continuous development of cancer treatment methods and technologies, the situation has improved [5]. But the problems of drug resistance and immunotoxicity cannot be ignored. Therefore, seeking new treatment targets and exploring their related mechanisms in the occurrence and development of lung cancer are the current research focus and urgent problem to be solved. Therefore, seeking new treatment targets and exploring their related mechanisms in the occurrence and development of lung cancer are the current research focus and urgent problem to be solved.
INSR and IRS-1 can mediate tumorigenesis and development as key mediators in the insulin signal transduction pathway, but their specific biological roles have not been fully clarified. Zhang et al. [6] found that downregulation of INSR can inhibit tumor cell proliferation, angiogenesis, lymphangiogenesis, and metastasis. Kim et al. [7] studied the effect of INSR expression on the survival of patients with NSCLC for the first time and found that INSR expression could be used as an independent prognostic factor for OS and RFS in patients with surgically resected early NSCLC. INSR has been proved to be abnormally expressed in a variety of tumor tissues, such as breast cancer, lung cancer, and gastric cancer [7–9]. Li et al. [10] first found that overexpression of IRS-1 can promote the malignant transformation of mouse embryonic fibroblasts. Subsequently, Tanaka et al. [11] further confirmed that IRS-1 overexpression can induce malignant transformation and proliferation of NIH3T3 fibroblasts by activating ERK1/2 pathway under low serum conditions. In addition, fibroblasts transfected with IRS can form colonies in soft agar, which is also highly tumorigenic when injected into nude mice. As a transmembrane protein, PD-L1 is considered to be an inhibitor of immune responses. It can combine with PD-1 to mediate the occurrence of tumor immune evasion [12]. With the approval of immune checkpoint inhibitors (ICIS), the treatment of immune checkpoints based on regulating PD-1/PD-L1 signaling pathway provides more options for the treatment of patients with advanced NSCLC [13, 14]. Gene expression studies indicate that PD-L1 expression is a biomarker of improved survival and response to immune checkpoint blockade therapy [15, 16]. IRS-1 expression is also associated with prognosis [17].
However, there are few reports on the correlation between INSR, IRS-1, and PD-L1 in NSCLC patients. Therefore, it is of great significance to explore the correlation between the expression levels of INSR, IRS-1, and PD-L1 and reveal the potential mechanism of insulin pathway-related receptors in the occurrence and development of lung cancer.
2. Materials and Methods
2.1. Tissue Samples
45 patients with NSCLC diagnosed in the Second Affiliated Hospital of Shandong First Medical University from June 2019 to August 2021 were selected. The 45 patients with nonsmall cell lung cancer, including 26 males and 19 females, did not receive radiotherapy, chemotherapy, and other antitumor treatments before surgery. The clinical data of all patients included in the experiment were complete, and the postoperative pathology had confirmed NSCLC. The TNM stage was determined according to the 2018 AJCC staging guidelines. This experiment has been approved by the medical ethics committee of our hospital before it is started.
2.2. Immunohistochemistry
The expression of INSR, IRS-1, and PD-L1 in 45 NSCLC tissues and 30 adjacent normal tissues were detected by immunohistochemistry (IHC). The results of immunohistochemical staining were determined by two pathologists using semiquantitative analysis method. The positive staining of INSR was localized in the cell membrane and cytoplasm, while the positive staining of IRS-1 was localized in the cytoplasm and nucleus. Observed the staining intensity and count the proportion of positive cells under a high power microscope. The intensity of the immunostaining was scored as follows: (a) no staining, 0; (b) pale yellow, 1; (c) brown-yellow, 2; and (d) brown, 3. The percentage of positive tumor cells was scored as (a) <5%, 0; (b) 5-25%, 1; (c) 26-50%, 2; (d) 51-75%, 3; and (e) >75%, 4. The two scores were multiplied to produce a weighted score for each case. Cases with weighted scores of more than 3 were defined as positive; otherwise; they were defined as negative. The positive staining of PD-L1 was mainly localized in the cell membrane, which was scored according to the proportion of tumor pigmented cells (TPS Score). When
2.3. Statistical Analysis
SPSS26.0 statistical software was used for data processing, and the comparison between positive expression rate, expression of each indicator, and clinicopathological data among groups was performed by chi-square test; the correlation between INSR, IRS-1, and PD-L1 expression was tested by Spearman rank correlation analysis. When the default
3. Results
3.1. Expression of INSR and IRS-1 in NSCLC and Adjacent Normal Tissues
The expression of INSR and IRS-1 in 45 cases of nonsmall cell lung cancer and 30 cases of adjacent normal lung tissues was detected by immunohistochemical staining (Figure 1). The results showed that INSR and IRS-1 are expressed in both lung cancer and adjacent normal tissues. The expression of INSR was dramatically higher in 29 (64.4%) cases compared with 11 (36.7%) cases in adjacent normal tissues (
[figure(s) omitted; refer to PDF]
Table 1
Difference of expression of INSR and IRS-1 in lung cancer and normal lung tissue.
Variable | Total | INSR (%) | IRS-1 (%) | ||||||
+ | - | + | - | ||||||
Tumor tissues | 45 | 29 (64.4) | 16 (35.6) | 5.580 | 0.018 | 26 (57.8) | 19 (42.2) | 4.309 | 0.038 |
Adjacent tissues | 30 | 11 (36.7) | 19 (63.3) | 10 (33.3) | 20 (66.7) |
3.2. The Relationship between INSR, IRS-1 Expression, and Clinicopathological Features in NSCLC
The results of statistical analysis showed that the positive expression of INSR was correlated with the degree of pathological differentiation of NSCLC patients, and the difference was statistically significant (
Table 2
The relationship between INSR, IRS-1 expression, and clinicopathological features in NSCLC.
Clinicopathological features | Total | INSR (%) | IRS-1 (%) | |||||
+ | - | + | - | |||||
Gender | Man | 26 | 17 (65.4) | 9 (34.6) | 0.878 | 14 (33.3) | 12 (33.3) | 0.532 |
Female | 19 | 12 (63.2) | 7 (36.8) | 12 (63.2) | 7 (36.8) | |||
Age | <60 | 20 | 13 (65.0) | 7 (35.0) | 0.944 | 12 (60.0) | 8 (40.0) | 0.787 |
≥60 | 25 | 16 (64.0) | 9 (36.0) | 14 (56.0) | 11 (44.0) | |||
Pathological type | Squamous carcinoma | 12 | 7 (58.3) | 5 (41.7) | 0.606 | 5 (41.7) | 7 (58.3) | 0.187 |
Adenocarcinoma | 33 | 22 (66.7) | 11 (33.3) | 21 (63.6) | 12 (36.4) | |||
TNM stage | 19 | 10 (52.6) | 9 (47.4) | 0.157 | 12 (63.2) | 7 (36.8) | 0.532 | |
26 | 19 (73.1) | 7 (26.9) | 14 (53.8) | 12 (46.2) | ||||
Tissue differentiation | Moderately-well | 27 | 14 (51.9) | 13 (48.1) | 0.031 | 16 (59.3) | 11 (40.7) | 0.805 |
Poorly | 18 | 15 (83.3) | 3 (16.7) | 10 (55.6) | 8 (44.4) | |||
Lymph node metastasis | No | 18 | 11 (61.1) | 7 (38.9) | 0.712 | 10 (55.6) | 8 (44.4) | 0.309 |
Yes | 27 | 15 (55.6) | 12 (44.4) | 19 (70.4) | 8 (29.6) |
3.3. The Relationship between PD-L1 Expression and Clinicopathological Features in NSCLC
There were 24 patients with positive expression, including 6 patients with high expression of PD-L1 (
Table 3
The relationship between PD-L1 expression and clinicopathological features in NSCLC.
Clinicopathological features | Total | PD-L1 (%) | |||
+ | - | ||||
Gender | Man | 26 | 14 (53.8) | 12 (46.2) | 0.936 |
Female | 19 | 10 (52.6) | 9 (47.4) | ||
Age | <60 | 20 | 12 (60.0) | 8 (40.0) | 0.423 |
≥60 | 25 | 12 (48.0) | 13 (52.0) | ||
Pathological type | Squamous carcinoma | 12 | 8 (66.7) | 4 (33.3) | 0.280 |
Adenocarcinoma | 33 | 16 (48.5) | 17 (51.5) | ||
TNM stage | 19 | 7 (36.8) | 12 (63.2) | 0.058 | |
26 | 17 (65.4) | 9 (34.6) | |||
Tissue differentiation | Moderately-well | 27 | 14 (51.9) | 13 (48.1) | 0.807 |
Poorly | 18 | 10 (55.6) | 8 (44.4) | ||
Lymph node metastasis | No | 18 | 6 (33.3) | 12 (66.7) | 0.028 |
Yes | 27 | 18 (66.7) | 9 (33.3) |
3.4. Correlation between INSR and PD-L1 Expressions in NSCLC
Spearman rank correlation was used to analyze the correlation between INSR, IRS-1, and PD-L1 expressions in nonsmall cell lung cancer. The results showed that there was no correlation between the expression of INSR and PD-L1 in nonsmall cell lung cancer (
Table 4
Correlation between INSR and PD-L1 expression in NSCLC.
INSR | PD-L1 | ||||
+ | - | ||||
+ | 17 | 12 | 29 | 0.143 | 0.350 |
- | 7 | 9 | 16 | ||
24 | 21 | 45 |
Table 5
Correlation between IRS-1 and PD-L1 expression in NSCLC.
IRS-1 | PD-L1 | ||||
+ | - | ||||
+ | 18 | 8 | 26 | 0.373 | 0.012 |
- | 6 | 13 | 19 | ||
24 | 21 | 45 |
4. Discussion
The reprogramming of tumor energy metabolism is considered to be one of the typical features of cancer, which endows tumor cells with the potential to continue to grow and proliferate in the tumor microenvironment (TME) with nutrient deficiency [18, 19]. As early as 1924, Ott Warburg first observed this abnormality of energy metabolism of tumor cells. Even in the case of sufficient oxygen supply, tumor cells still prefer to change their metabolic dependence from mitochondrial oxidative phosphorylation (OXPHOS) to glycolysis to generate energy [20]. After that, Zhou et al. [21] also found that drug-resistant cell lines have higher levels of aerobic glycolysis capacity, suggesting that there is a biochemical link between drug resistance and glycolysis. Oronsky et al. [22] further proposed that ATP produced by intracellular metabolism is the key determinant of chemoradiotherapy resistance. Controlling the metabolic process in vivo to limit ATP level can improve chemoradiotherapy sensitivity. In conclusion, energy metabolism reprogramming plays an important role in tumorigenesis and development. Therefore, targeted intervention on glycolysis has brought a new idea for tumor treatment, and the relevant studies have also confirmed the effectiveness of this treatment method [23]. Glucose transmembrane transport is considered as the rate limiting step of glycolysis [24]. In order to make up for the low efficiency of glycolysis energy production, tumor cells tend to ingest glucose at a higher rate, which is mediated by glucose transporters (GLUTs). As a transmembrane protein, gluts are widely distributed in human tissues. Up to now, 14 glut subtypes (Glut 1-14) have been found. Gluts, especially Glut1/3, have been observed to be highly expressed in a variety of different types of tumors, such as lung cancer and pancreatic cancer. Its overexpression not only significantly enhanced glucose uptake but also showed a close relationship with the poor tumor prognosis [24–26]. Small molecule inhibitors targeting gluts, including phloretin, WZB117, and STF-31, had also shown good effects in antitumor therapy [27, 28]. Studies have shown that phosphoinositide-3 kinase (PI3K/AKT) and other pathways are central regulators of glycolysis, tumor metabolism, and cancer cell proliferation [29]. In tumor cells, PI3K/AKT pathway can induce the expression of gluts and enhance the glycolysis process [30–33]. In addition, PI3K-Akt pathway is also a classical insulin downstream signaling pathway. In the insulin signaling pathway, when insulin binds to the INSRs on the cell membrane, it enhances the binding affinity between the receptor and IRS protein, causing the phosphorylation of IRSs to be activated. Then, IRSs can activate this pathway by recruiting and activating PI3K. Therefore, we have reason to believe that INSR and IRS-1 are involved in the process of glycolysis metabolism and play an important role in maintaining the survival and proliferation of tumor cells. They are expected to become potential targets for antitumor therapy. Our studies also showed that the positive rate of INSR and IRS-1 protein expression in NSCLC tissues was significantly higher than that in adjacent normal tissues (
PD-L1 is a recognized immunosuppressive molecule. Tumor cells can negatively regulate the activity of T cells by increasing the expression of PD-L1 on the cell surface, so as to achieve the occurrence of immune escape. With the approval of immune checkpoint inhibitor, immunotherapy against PD-L1 has shown good therapeutic effect in nonsmall cell lung cancer, but its immune-related adverse toxicity cannot be ignored. Therefore, looking for new targets to specifically regulate PD-L1 expression will help to more accurately block tumor-related PD-1/PD-L1 pathway and reduce adverse immune reactions. Previous studies have shown that PI3K/AKT pathway activation can promote PD-L1 expression by increasing exogenous signals or reducing the expression of negative regulatory factors such as PTEN [36]. Zhao et al. [37] confirmed that PD-1/PD-L1 blockade may inhibit the apoptosis of CD8+ T cells in gastrointestinal stromal tumors (GIST) by regulating PI3K/Akt/mTOR pathway. Stutvoet et al. [38] showed that inhibition of MAPK pathway can regulate EGF- and IFN-induced PD-L1 expression in lung adenocarcinoma. These results all suggest that the expression of PD-L1 can be regulated by PI3K/AKT and MAPK signaling pathway. These two pathways happen to be the most important downstream pathways of INSR and IRS-1. Whether they can regulate the abnormal expression of PD-L1 through these pathways has not been clarified yet. IRS proteins play a key role in tumor metabolism by regulating signaling of INSR. However, IRS-1 is also ubiquitously expressed in cancer cells. The differential expressions of IRS-1 versus IRS-2 may regulate drug responses. Further studies are essential to clarify complex signaling mechanisms [39]. Therefore, we used spearman correlation analysis to explore the relationships between them. The result showed that there is a positive correlation between the expression of IRS-1 and PD-L in NSCLC tissues (
5. Conclusion
INSR and IRS-1 are both overexpression in lung cancer tissues, and IRS-1 expression had a significant positive correlation with PD-L1 expression. The expression of IRS-1 may further mediate the occurrence of tumor immune escape. Therefore, a better understanding of the regulatory mechanism of IRS-1 on PD-L1 expression will help to find new targets for NSCLC immunotherapy and provide new clinical ideas for immunosuppressive therapy.
Authors’ Contributions
Ting Ma (email:
Acknowledgments
This study was supported by Shandong Province Medical and Health Science and Technology Development Plan (Grant no.: 2019WS400).
[1] Z. C. Liu, Z. X. Li, Y. Zhang, "Interpretation of 2020 global cancer statistical report," Journal of Multidisciplinary Cancer Management, vol. 7 no. 2, 2021.
[2] A. K. Ganti, A. B. Klein, I. Cotarla, B. Seal, E. Chou, "Update of incidence, prevalence, survival, and initial treatment in patients with non–small cell lung cancer in the US," JAMA Oncology, vol. 7 no. 12, pp. 1824-1832, DOI: 10.1001/jamaoncol.2021.4932, 2021.
[3] W. D. Travis, E. Brambilla, G. J. Riely, "New pathologic classification of lung cancer: relevance for clinical practice and clinical trials," Journal of Clinical Oncology, vol. 31 no. 8, pp. 992-1001, DOI: 10.1200/JCO.2012.46.9270, 2013.
[4] C. Allemani, T. Matsuda, V. Di Carlo, R. Harewood, M. Matz, M. Nikšić, A. Bonaventure, M. Valkov, C. J. Johnson, J. Estève, O. J. Ogunbiyi, "Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries," The Lancet, vol. 391 no. 10125, pp. 1023-1075, DOI: 10.1016/S0140-6736(17)33326-3, 2018.
[5] R. L. Siegel, K. D. Miller, H. E. Fuchs, A. Jemal, "Cancer statistics, 2022," CA: a Cancer Journal for Clinicians, vol. 72 no. 1,DOI: 10.3322/caac.21708, 2022.
[6] H. Zhang, D. H. Fagan, X. Zeng, K. T. Freeman, D. Sachdev, D. Yee, "Inhibition of cancer cell proliferation and metastasis by insulin receptor downregulation," Oncogene, vol. 29 no. 17, pp. 2517-2527, DOI: 10.1038/onc.2010.17, 2010.
[7] J. S. Kim, E. S. Kim, D. Liu, J. J. Lee, L. Solis, C. Behrens, S. M. Lippman, W. K. Hong, I. I. Wistuba, H. Y. Lee, "Prognostic impact of insulin receptor expression on survival of patients with nonsmall cell lung cancer," Cancer, vol. 118 no. 9, pp. 2454-2465, DOI: 10.1002/cncr.26492, 2012.
[8] J. H. Law, G. Habibi, K. Hu, H. Masoudi, M. Y. C. Wang, A. L. Stratford, E. Park, J. M. W. Gee, P. Finlay, H. E. Jones, R. I. Nicholson, J. Carboni, M. Gottardis, M. Pollak, S. E. Dunn, "Phosphorylated insulin-like growth factor-i/insulin receptor is present in all breast cancer subtypes and is related to poor survival," Cancer Research, vol. 68 no. 24, pp. 10238-10246, DOI: 10.1158/0008-5472.CAN-08-2755, 2008.
[9] M. Saisana, S. M. Griffin, F. E. B. May, "Insulin and the insulin receptor collaborate to promote human gastric cancer," Gastric Cancer, vol. 25 no. 1, pp. 107-123, DOI: 10.1007/s10120-021-01236-y, 2022.
[10] L. Li, Y. Liang, L. Kang, Y. Liu, S. Gao, S. Chen, Y. Li, W. You, Q. Dong, T. Hong, Z. Yan, S. Jin, T. Wang, W. Zhao, H. Mai, J. Huang, X. Han, Q. Ji, Q. Song, C. Yang, S. Zhao, X. Xu, Q. Ye, "Transcriptional regulation of the Warburg effect in cancer by SIX1," Cancer Cell, vol. 33 no. 3, pp. 368-385.e7, DOI: 10.1016/j.ccell.2018.01.010, 2018.
[11] S. Tanaka, T. Ito, J. R. Wands, "Neoplastic transformation induced by insulin receptor substrate-1 overexpression requires an interaction with both Grb2 and Syp signaling molecules ∗," Journal of Biological Chemistry, vol. 271 no. 24, pp. 14610-14616, DOI: 10.1074/jbc.271.24.14610, 1996.
[12] L. Ai, A. Xu, J. Xu, "Roles of PD-1/PD-L1 pathway: signaling, cancer, and beyond," Regulation of Cancer Immune Checkpoints, vol. 1248, pp. 33-59, DOI: 10.1007/978-981-15-3266-5_3, 2020.
[13] K. C. Ohaegbulam, A. Assal, E. Lazar-Molnar, Y. Yao, X. Zang, "Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway," Trends in Molecular Medicine, vol. 21 no. 1, pp. 24-33, DOI: 10.1016/j.molmed.2014.10.009, 2015.
[14] J. Qu, Q. Mei, L. Liu, T. Cheng, P. Wang, L. Chen, J. Zhou, "The progress and challenge of anti-PD-1/PD-L1 immunotherapy in treating non-small cell lung cancer," Therapeutic Advances in Medical Oncology, vol. 13,DOI: 10.1177/1758835921992968, 2021.
[15] P. N. Aguiar, R. A. De Mello, P. Hall, H. Tadokoro, G. D. Lima Lopes, "PD-L1 expression as a predictive biomarker in advanced non-small-cell lung cancer: updated survival data," Immunotherapy, vol. 9 no. 6, pp. 499-506, DOI: 10.2217/imt-2016-0150, 2017.
[16] C. Wang, T. Lu, R. Xu, X. Chang, S. Luo, B. Peng, J. Wang, L. Yao, K. Wang, Z. Shen, J. Zhao, L. Zhang, "A bioinformatics-based immune-related prognostic index for lung adenocarcinoma that predicts patient response to immunotherapy and common treatments," Journal of Thoracic Disease., vol. 14 no. 6, pp. 2131-2146, DOI: 10.21037/jtd-22-494, 2022.
[17] A. J. Piper, J. L. Clark, J. Mercado-Matos, A. N. Matthew-Onabanjo, C. C. Hsieh, A. Akalin, L. M. Shaw, "Insulin receptor substrate-1 (IRS-1) and IRS-2 expression levels are associated with prognosis in non-small cell lung cancer (NSCLC)," PLoS One, vol. 14 no. 8, article e0220567,DOI: 10.1371/journal.pone.0220567, 2019.
[18] D. Hanahan, R. A. Weinberg, "Hallmarks of cancer: the next generation," Cell, vol. 144 no. 5, pp. 646-674, DOI: 10.1016/j.cell.2011.02.013, 2011.
[19] R. J. DeBerardinis, N. S. Chandel, "Fundamentals of cancer metabolism," Science Advances, vol. 2 no. 5, article e1600200,DOI: 10.1126/sciadv.1600200, 2016.
[20] P. Vaupel, H. Schmidberger, A. Mayer, "The Warburg effect: essential part of metabolic reprogramming and central contributor to cancer progression," International Journal of Radiation Biology, vol. 95 no. 7, pp. 912-919, DOI: 10.1080/09553002.2019.1589653, 2019.
[21] Y. Zhou, F. Tozzi, J. Chen, F. Fan, L. Xia, J. Wang, G. Gao, A. Zhang, X. Xia, H. Brasher, W. Widger, L. M. Ellis, Z. Weihua, "Intracellular ATP levels are a pivotal determinant of chemoresistance in colon cancer cells," Cancer Research, vol. 72 no. 1, pp. 304-314, DOI: 10.1158/0008-5472.CAN-11-1674, 2012.
[22] T. Oronsky, N. Oronsky, G. Fanger, C. Parker, S. Caroen, M. Lybeck, J. Scicinski, "Follow the ATP: tumor energy production: a perspective," Anti-Cancer Agents in Medicinal Chemistry, vol. 14 no. 9, pp. 1187-1198, DOI: 10.2174/1871520614666140804224637, 2014.
[23] Y. Zhao, E. B. Butler, M. Tan, "Targeting cellular metabolism to improve cancer therapeutics," Cell Death & Disease, vol. 4 no. 3, pp. e532-e532, DOI: 10.1038/cddis.2013.60, 2013.
[24] C. C. Barron, P. J. Bilan, T. Tsakiridis, E. Tsiani, "Facilitative glucose transporters: implications for cancer detection, prognosis and treatment," Metabolism, vol. 65 no. 2, pp. 124-139, DOI: 10.1016/j.metabol.2015.10.007, 2016.
[25] J. Y. Sung, G. Y. Kim, S. J. Lim, Y. K. Park, Y. W. Kim, "Expression of the GLUT1 glucose transporter and p53 in carcinomas of the pancreatobiliary tract," Pathology-Research and Practice, vol. 206 no. 1, pp. 24-29, DOI: 10.1016/j.prp.2009.07.018, 2010.
[26] C. Yin, B. Gao, J. Yang, J. Wu, "Glucose transporter-1 (GLUT-1) expression is associated with tumor size and poor prognosis in locally advanced gastric cancer," Medical Science Monitor Basic Research, vol. 26, article e920778,DOI: 10.12659/MSMBR.920778, 2020.
[27] D. Kraus, J. Reckenbeil, N. Veit, S. Kuerpig, M. Meisenheimer, I. Beier, H. Stark, J. Winter, R. Probstmeier, "Targeting glucose transport and the NAD pathway in tumor cells with STF-31: a re-evaluation," Cellular Oncology, vol. 41 no. 5, pp. 485-494, DOI: 10.1007/s13402-018-0385-5, 2018.
[28] E. B. Butler, Y. Zhao, C. Muñoz-Pinedo, J. Lu, M. Tan, "Stalling the engine of resistance: targeting cancer metabolism to overcome therapeutic resistance," Cancer Research, vol. 73 no. 9, pp. 2709-2717, DOI: 10.1158/0008-5472.CAN-12-3009, 2013.
[29] R. Courtnay, D. C. Ngo, N. Malik, K. Ververis, S. M. Tortorella, T. C. Karagiannis, "Cancer metabolism and the Warburg effect: the role of HIF-1 and PI3K," Molecular Biology Reports, vol. 42 no. 4, pp. 841-851, DOI: 10.1007/s11033-015-3858-x, 2015.
[30] A. Barthel, S. T. Okino, J. Liao, K. Nakatani, J. Li, J. P. Whitlock, R. A. Roth, "Regulation of GLUT1 gene transcription by the serine/threonine kinase Akt1 ∗," Journal of Biological Chemistry, vol. 274 no. 29, pp. 20281-20286, DOI: 10.1074/jbc.274.29.20281, 1999.
[31] H. M. Wang, Y. J. Lu, L. He, N. J. Gu, S. Y. Wang, X. S. Qiu, E. H. Wang, G. P. Wu, "HPV16 E6/E7 promote the translocation and glucose uptake of GLUT1 by PI3K/AKT pathwayviarelieving miR-451 inhibitory effect on CAB39 in lung cancer cells," Therapeutic Advances in Chronic Disease, vol. 11,DOI: 10.1177/2040622320957143, 2020.
[32] J. Yu, J. Li, S. Zhang, X. Xu, M. Zheng, G. Jiang, F. Li, "IGF-1 induces hypoxia-inducible factor 1 α -mediated GLUT3 expression through PI3K/Akt/mTOR dependent pathways in PC12 cells," Brain Research, vol. 1430, pp. 18-24, DOI: 10.1016/j.brainres.2011.10.046, 2012.
[33] J. Jiang, H. Y. Ren, G. J. Geng, Y. J. Mi, Y. Liu, N. Li, S. Y. Yang, D. Y. Shen, "Oncogenic activity of insulin in the development of non-small cell lung carcinoma," Oncology Letters, vol. 15 no. 1, pp. 447-452, DOI: 10.3892/ol.2017.7347, 2018.
[34] S. M. Heckl, F. Mau, A. Senftleben, T. Daunke, S. Beckinger, S. Abdullazade, S. Schreiber, C. Röcken, S. Sebens, H. Schäfer, "Programmed death-ligand 1 (PD-L1) expression is induced by insulin in pancreatic ductal adenocarcinoma cells pointing to its role in immune checkpoint control," Medical Science, vol. 9 no. 3, 2021.
[35] X. Liu, K. Yamaguchi, K. Takane, C. Zhu, M. Hirata, Y. Hikiba, S. Maeda, Y. Furukawa, T. Ikenoue, "Cancer-associated IDH mutations induce Glut1 expression and glucose metabolic disorders through a PI3K/Akt/mTORC1-Hif1 α axis," PLoS One, vol. 16 no. 9, article e0257090,DOI: 10.1371/journal.pone.0257090, 2021.
[36] J. Chen, C. C. Jiang, L. Jin, X. D. Zhang, "Regulation of PD-L1: a novel role of pro-survival signalling in cancer," Annals of Oncology, vol. 27 no. 3, pp. 409-416, DOI: 10.1093/annonc/mdv615, 2016.
[37] R. Zhao, Y. Song, Y. Wang, Y. Huang, Z. Li, Y. Cui, M. Yi, L. Xia, W. Zhuang, X. Wu, Y. Zhou, "PD-1/PD-L1 blockade rescue exhausted CD8+ T cells in gastrointestinal stromal tumours via the PI3K/Akt/mTOR signalling pathway," Cell Proliferation, vol. 52 no. 3, article e12571,DOI: 10.1111/cpr.12571, 2019.
[38] T. S. Stutvoet, A. Kol, E. G. E. de Vries, M. Bruyn, R. S. N. Fehrmann, A. G. T. Terwisscha van Scheltinga, S. Jong, "MAPK pathway activity plays a key role in PD-L1 expression of lung adenocarcinoma cells," The Journal of Pathology, vol. 249 no. 1, pp. 52-64, DOI: 10.1002/path.5280, 2019.
[39] L. M. Shaw, "The insulin receptor substrate (IRS) proteins: at the intersection of metabolism and cancer," Cell Cycle, vol. 10 no. 11, pp. 1750-1756, DOI: 10.4161/cc.10.11.15824, 2011.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2022 Ma Ting et al. This work is licensed under http://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Objective. To study the expression and correlation of insulin receptor (INSR), insulin receptor substrate-1 (IRS-1), and programmed cell death ligand-1 (PD-L1) in nonsmall cell lung cancer (NSCLC). Methods. 45 lung cancer tissues and 30 adjacent normal tissues of NSCLC patients diagnosed in the Second Affiliated Hospital of Shandong First Medical University from June 2019 to August 2020 were selected. The expressions of INSR, IRS-1, and PD-L1 proteins in tumor tissues and adjacent tissues of NSCLC were detected by immunohistochemical staining. Results. The expression of INSR and IRS-1 in NSCLC was significantly higher than that in adjacent normal lung tissue (
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details



1 Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, 271000 Shandong Province, China; The Second Affiliated Hospital of Shandong First Medical University, Tai’an, 271000 Shandong Province, China
2 Department of Chemotherapy, The Second Affiliated Hospital of Shandong First Medical University, Tai’an 271000, Shandong Province, China
3 Basic Medical College, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, 271000 Shandong Province, China
4 Department of Radiation Oncology, The Second Affiliated Hospital of Shandong First Medical University, Tai’an 271000, Shandong Province, China
5 The Second Affiliated Hospital of Shandong First Medical University, Tai’an, 271000 Shandong Province, China