Open Access

Activation of Janus kinase 1 confers poor prognosis in patients with non‑small cell lung cancer

  • Authors:
    • Dan Liu
    • Yi Huang
    • Li Zhang
    • Dong‑Ni Liang
    • Li Li
  • View Affiliations

  • Published online on: August 1, 2017     https://doi.org/10.3892/ol.2017.6690
  • Pages: 3959-3966
  • Copyright: © Liu 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

The activation of Janus kinase 1 (JAK1) has been reported to occur in non‑small cell lung cancer (NSCLC), activating the JAK/signal transducers and activators of transcription cascade. However, the association between JAK1 activation and the prognostic value in NSCLC remains unclear. The present study initially investigated the association between expression of the activated form of JAK1 (p‑JAK1) and prognosis in patients with NSCLC. A cohort of 142 resected primary NSCLC tissue samples, including 74 adenocarcinoma (ADCC) and 68 squamous cell carcinoma samples, were analyzed. p‑JAK1 expression status was determined by immunohistochemistry. Evaluation of epidermal growth factor receptor (EGFR) gene amplification by fluorescence in situ hybridization was subsequently performed in 74 ADCC samples. The prognostic significance of p‑JAK1 expression and EGFR gene amplification were evaluated with univariate and multivariate survival analyses. Compared with normal lung tissue, p‑JAK1 expression level was significantly increased in NSCLC (P<0.001). Positive p‑JAK1 expression indicated a poor prognosis, particularly for patients in early stages (stage I/II, including tumor size <3 cm, Lymph node invasion N0/1; all P<0.05). p‑JAK1 expression was an independent predictor of a poor prognosis (P=0.022). The overall survival time for patients with positive p‑JAK1 expression and EGFR‑amplified tumors was significantly shortened compared with patients with tumors negative for one or both features (both features present vs. neither feature present, P<0.001). The results provided clinical evidence that the activation of JAK1 was an independent prognostic factor, particularly in early stage NSCLC. The combination of EGFR gene amplification and p‑JAK1 expression may be a novel target for the selection of individual therapy strategies and predicting the effects of therapy for NSCLC.

Introduction

Lung cancer is one of the leading causes of cancer-associated mortality, accounting for 27% (including 26% for females, and 28% for males), and non-small cell lung cancer (NSCLC) accounts for 80–85% of all lung cancer-associated mortalities (1,2). Despite advances in the understanding of the molecular mechanisms of lung cancer and the development of novel chemotherapeutic agents, the 5-year survival rates for lung and bronchus cancer remained <18% from 2004 to 2010 (2). A number of studies have focused on progressing the understanding of oncogenic kinase signaling pathways, which has provided targets for developing effective therapeutic strategies in order to improve clinical outcomes (3).

One of the potential candidates for therapy is the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. JAK/STAT is one of the pleiotropic cascades that may transduce a multitude of signals for development and homeostasis in animals, from humans to flies (4). JAK family members have been reported to be dysregulated in malignant tumors, including in colorectal, prostate and myeloproliferative cancer (58). The mammalian JAK family includes four members: JAK1, JAK2, JAK3 and Tyk2. All JAKs exhibit broad patterns of expression with the exception of JAK3, which is restricted to leukocytes (9).

JAK1 binds to various cytokines non-covalently to mediate cell proliferation and differentiation (10). JAK1 knockout mice die perinatally (9). JAK1 mutation has been reported in hepatocellular carcinoma, acute lymphoblastic leukemia, lung and gastric cancer (10,11). Phosphorylated (p)-JAK1, the active form of JAK1, mediates the phosphorylation of receptors and the major substrates for JAK family members, STATs (4). For example, p-JAK1 expression was detected in primary esophageal squamous cell carcinoma and not in normal esophageal squamous cells. p-JAK1 expression was associated with a reduced overall survival time (12). Additionally, a previous study by the present authors demonstrated that JAK1 expression was significantly increased in NSCLC clinical samples compared with normal samples (P>0.001), while p-JAK1 (Tyk 1022) showed trends of positive expression, though these did not reach statistical significance (P=0.055), potentially owing to a small sample size (13). This indicated that JAK1 activation was abnormal in NSCLC.

Lung adenocarcinoma (ADCC) is the most common histological subtype of NSCLC. Patients with ADCC and a high epidermal growth factor receptor (EGFR) copy number can be treated with EGFR-tyrosine kinase inhibitors (14,15). EGFR gene amplification has also been reported to be associated with prognosis of lung cancer, though there is some controversy in this regard (16,17). However, the combination of EGFR gene amplification and JAK1 activation for predicting cancer prognosis has not been extensively studied. This has incited the present study, which will investigate associations between JAK1 activation, EGFR gene amplification and survival status in patients with NSCLC.

Materials and methods

Tissue collection

The study cohort consisted of 142 patients (40 female and 102 male) with a median age of 63 years (range, 20–84 years). A total of 142 paraffin-embedded resected primary NSCLC samples were analyzed from the archives of the Pathology Department at Sichuan Provincial People's Hospital (Chengdu, China) from December 2004 to February 2007, including 74 cases of ADCC and 68 cases of squamous cell carcinoma (SqCC) A total of 142 adjacent normal pulmonary tissue specimens were also resected in the same tissue blocks. Staging was performed according to the International Union Against Cancer's tumor-node-metastasis system (18). Differentiation and histological type were scored according to the World Health Organization classification for NSCLC (19). None of the patients had received neoadjuvant therapy prior to surgical resection. Following the surgery, the patients underwent standard therapy procedure, according to the National Comprehensive Cancer Network Clinical Practice Guideline for Oncology, NSCLC, 2004 (20). Informed consent was obtained from all individuals included in the present study. Institutional review board approval for the study was obtained from Sichuan Provincial People's Hospital.

Immunohistochemical staining (IHC)

IHC staining was performed as previously described (3). Briefly, the 4-µm sections underwent deparaffinization, hydration and endogenous peroxide blocking. Antigen retrieval was performed by heating at 95°C for 30 min in Tris/ethylenediaminetetraacetic acid retrieval solution. The sections were blocked with 3% bovine serum albumin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and incubated with primary p-JAK1 antibody (cat. no. 11149, 1:100 dilution; Signalway Antibody LLC., College Park, MY, ML, USA) overnight at 4°C. The membranes were subsequently incubated with enough secondary peroxidase-labeled polymer-conjugated goat anti-rabbit antibody to cover the specimen (cat. no. K4007; undiluted; EnVsion; Dako; Agilent Technologies, Inc., Santa Clara, CA, USA), according to the manufacturer's protocol, for 30 min at room temperature. For staining, 3,3-daiminobenzidine chromogen was applied. Finally, the sections were counterstained with hematoxylin for 3 min at room temperature, fixed and mounted.

Scoring for immunohistochemical staining

Scoring for immunochemical staining was evaluated as previously described (3) by two independent pathologists. Briefly, the expression of p-JAK1 was assessed semi-quantitatively on the basis of criteria that accounted for the fraction and intensity of immunostaining of the tumor cells. The fraction score was defined as: 0 (0%), 1 (<20%), 2 (20–50%) and 3 (>50%). The intensity of p-JAK1 staining was scored as 0 (no appreciable staining), 1 (barely detectable staining), 2 (readily identifiable brown staining) and 3 (dark brown staining). A total score was calculated by multiplying the fraction and the intensity score. A tumor sample was considered positive if the score was ≥4, and negative otherwise.

Determination of EGFR gene amplification by fluorescence in-situ hybridization (FISH)

EGFR FISH analysis was performed with the dual-color EGFR SpectrumOrange/CEP7 SpectrumGreen probe and paraffin pretreatment reagent kit (both from Vysis; cat. nos. 05J48-001 and 32–801210, respectively; Abbott Laboratories, Chicago, IL, USA) as previously described (21). Briefly, the paraffin sections were deparaffinized, dehydrated and digested with protease K. The slides were denatured by heating to 75°C for 5 min and dehydrated in ethanol. The probes were denatured for 5 min at 75°C prior to hybridization. Each slide was hybridized at 37°C overnight and washed in 2X saline-sodium citrate buffer/0.3% NP40 at 72°C for 2 min. The nuclei were counterstained with DAPI/antifade 1 (Vysis; Abbott Laboratories, Chicago, IL, USA). A minimum of 100 non-overlapping tumor cell nuclei were scored in each case, according to the University of Colorado Cancer Center criteria (22). The BX-51/Genus FISH Imaging system (DP71/70/DP30BW software, DP-BSW Ver.03.02) was used for imaging (Olympus, Tokyo, Japan).

Statistical analysis

The association between clinical characteristics and p-JAK1 expression was determined using Pearson's χ2 test. When one of the expected values in a 2×2 table was <5, Fisher's exact test would be used. The Kaplan-Meier method was used for the univariate analysis of survival time between groups based on clinical characteristics and p-JAK1 expression. Multivariate analysis was performed using Cox regression model analysis. Only markers that were significant predictors in univariate analysis were included in the multivariate analysis. All tests were two-sided and P<0.05 was considered to indicate a statistically significant difference. SPSS 17.0 for Windows (SPSS Inc., Chicago, IL, USA) was used for these analyses.

Results

JAK1 expression in non-small cell lung cancer tissues

Representative IHC staining images of p-JAK1 expression in NSCLC and adjacent normal lung tissues are displayed in Fig. 1. Positive p-JAK1 expression was observed in the cytoplasm and nucleus, as indicated by arrows. In the NSCLC tissues, 56/142 cases (39.4%) exhibited positive p-JAK1 expression, whereas 11/142 cases (7.7%) were positive in the adjacent normal tissues (Table I). The difference was highly significant (P<0.001).

Table I.

p-JAK1 expression in NSCLC and adjacent normal tissues.

Table I.

p-JAK1 expression in NSCLC and adjacent normal tissues.

Parameterp-JAK1+, n (%)p-JAK1, n (%)P-valuea
NSCLC tissue56 (39.4)86 (60.6)<0.001
Normal control11 (7.7)131 (92.3)

a By Perason's χ2 test. p-JAK1, phosphorylated Janus kinase 1; NSCLC, non-small cell lung carcinoma.

Association between clinical characteristics and prognosis in patients with NSCLC

As listed in Table II, follow-up information was available for all patients. Survival time differed significantly by age (>60 vs. ≤60), lymph node invasion (N0/1 vs. N2/3) and stage (I/II vs. III/IV; P=0.005, P<0.001 and P=0.001, respectively). Gender, tumor size and histological type were not significantly associated with survival time.

Table II.

Clinical characteristics and prognosis of patients with non-small cell lung carcinoma.

Table II.

Clinical characteristics and prognosis of patients with non-small cell lung carcinoma.

ParameterPatients, nMedian survival time, range (months)P-valuea
Age 0.005
  <60 years5663 (3–94)
  ≥60 years8660 (1–96)
Gender 0.561
  Female4063 (3–96)
  Male10261 (1–94)
Histological type 0.959
  Adenocarcinoma7461 (3–96)
  Squamous cell carcinoma6862.5 (1–94)
Tumor size 0.446
  <3 cm5961.5 (6–96)
  ≥3 cm8360 (1–94)
Lymph node invasion <0.001
  N0/112263 (1–96)
  N2/32039 (3–67)
Distant metastasis 0.973
  M013861 (1–96)
  M1457.5 (41–85)
TNM stage 0.001
  I/II9262.5 (1–96)
  III/IV5051.5 (3–89)

a By log-rank test. TNM, tumor-node-metastasis staging system.

Association between clinical characteristics and p-JAK1 expression

Pearson's χ2 or Fisher's exact test were performed to assess the significance of associations between clinical characteristics and p-JAK1 expression. As shown in Table III, p-JAK1 expression was associated with histological type, as 29.7% of ADCC samples and 50.0% of SqCC samples were p-JAK1+ (P=0.014). p-JAK1 expression status was also associated with tumor size and stage (P=0.021 and P=0.009, respectively). Other clinical characteristics, including age, gender, lymph node invasion and distant metastasis, were not associated with p-JAK1 expression status (P>0.05).

Table III.

p-JAK1 expression and clinical characteristics in patients with non-small cell lung cancer.

Table III.

p-JAK1 expression and clinical characteristics in patients with non-small cell lung cancer.

Characteristic p-JAK1+ p-JAK1P-value
Age 0.867a
  <602235
  ≥603451
Gender 0.290a
  Female1327
  Male4359
Histological type 0.014a
  Adenocarcinoma2252
  Squamous cell carcinoma3434
Tumor size 0.029a
  <3 cm1742
  ≥3 cm3944
Lymph node invasion 0.297a
  N0/14676
  N2/31010
Distant metastasis 0.300b
  M05385
  M131
TNM stage 0.009a
  I/II2963
  III/IV2723

a By Perason's χ2 test;

b by Fisher's exact test. p-JAK1, phosphorylated Janus kinase 1; TNM, tumor-node-metastasis staging system.

Survival status and p-JAK1 expression

Positive p-JAK1 expression was associated with a poor prognosis in NSCLC (P=0.039; Fig. 2A-C). However when divided by histological type, the difference was only statistically significant in ADCC samples (P=0.007) and not in SqCC (P=0.612). Subjects with p-JAK1 expression demonstrated reduced survival time in early stage NSCLC, including patients with tumor size <3 cm, N0/1 and stage I/II (P=0.016, P=0.034 and P=0.048, respectively; Fig. 3A-F). The difference in survival time in patients with late stage disease, including tumor size ≥3 cm, N2/3 or stage III/IV was not significant.

Survival status and p-JAK1 and EGFR expression in ADCC

To investigate whether associations between JAK1 activation, EGFR gene amplification and prognosis in patients with ADCC were significant, EGFR status was examined with FISH analysis, and univariate survival analyses were performed for the 74 ADCC subjects (Table V). EGFR gene amplification was determined by FISH (Fig. 4), 29/74 ADCC cases exhibited EGFR gene amplification (designated as EGFR+). As shown in Table V, survival time for patients with p-JAK1+/EGFR+ was significantly reduced compared with those with p-JAK1/EGFR or p-JAK1/EGFR+ combinations (P<0.001 and P=0.004, respectively), but not the p-JAK1+/EGFR combination (P=0.052; Fig. 5). The results indicated that the EGFR amplification and p-JAK1+ combination may be a novel target to inform the selection of individual therapy strategies and for predicting the effect of therapy in ADCC.

Table V.

Effects of p-JAK1 expression and EGFR gene duplication status on the prognosis of patients with lung adenocarcinoma, as determined by Kaplan-Meier analysis.

Table V.

Effects of p-JAK1 expression and EGFR gene duplication status on the prognosis of patients with lung adenocarcinoma, as determined by Kaplan-Meier analysis.

StatusPatients, nMedian survival time (months) P-valuea
p-JAK1+/EGFR+716 (3–60)
p-JAK1/EGFR3061.5 (15–96)<0.001
p-JAK1/EGFR+2262 (13–94)0.004
p-JAK1+/EGFR1549 (12–90)0.052

a By log-rank test, compared with p-JAK1+/EGFR+ patients. p-JAK1, phosphorylated Janus kinase 1; EGFR, epidermal growth factor receptor.

Discussion

Phosphorylation of JAK1 is important for the activation of JAK/STAT pathways and their downstream cascades (10). In the present study, it was identified that the activation of JAK1 was associated with a poorer prognosis in NSCLC (P=0.039), particularly in ADCC (P=0.007). p-JAK1 was identified as an independent predictor for poor prognosis (P=0.022). Overall survival time for patients with p-JAK1+ and EGFR gene duplication was significantly reduced compared with patients with one or neither trait (P=0.001).

A previous study by the present authors and studies by others have indicated that inhibition of JAK signaling exhibits anticancer and anti-angiogenetic effects in human cancer lines and xenograft tumors, including lung cancer (13,2325). These results support the hypothesis that JAK1 activation is important for tumorigenesis in NSCLC. In the present study, when subdivided by histotype, differences in prognosis were identified as statistically significant. Positive p-JAK1 was an independent predictor for decreased survival times only for patients with ADCC, not SqCC. This finding suggests a specific role for JAK1 in lung ADCC. In several lung ADCC cell lines, JAK family inhibitor AZD1480 has demonstrated to be able to potently block STAT3 signaling and oncogenesis (24), which supports the results of the present study to some extent. However, the research on the effect of p-JAK1 expression on prognosis in lung SqCC has been limited. You et al (12) reported that JAK/STAT pathway activations were associated with shorter survival time in esophageal SqCC cases. As only 68 cases of SqCC patients were recruited to the present study, further evaluation should be performed using a larger amount of cases.

In the present study, it was further identified that tumor size <3 cm, N0/1 and stage I/II patients with positive p-JAK1 expression had shorter survival time (all P<0.05), whereas the difference was not significant for patients in advanced stages (Fig. 3). This finding indicated that the activation of JAK1 may be an early event in the tumorigenesis of NSCLC. Therefore, p-JAK1 could be a target for early diagnosis and prognosis prediction.

In the present study, patients with p-JAK1+ and EGFR duplication exhibited the poorest prognosis while patients with p-JAK1 and no EGFR duplication exhibited the most favorable prognosis (median survival time, 16 vs. 61.5 months). Although contradictory results were previously reported (14,15), EGFR FISH status alone was not associated with the patient survival times (data not shown). To date, few studies have focused on the interaction between EGFR and JAKs at the cellular and molecular level. However, it has been identified that crosstalk between the JAK/STAT and EGFR pathways mediates adenomatous polyposis coil 1-driven intestinal stem cell hyperplasia in drosophila (26). Furthermore, prolactin, a JAK2-coupled cytokine receptor, has also been demonstrated to synergistically augment EGF signaling in T47D breast cancer cells (27). In NSCLC, how EGFR and JAK1 interact to exert biological effects should be further elucidated.

The present study indicated that JAK1 activation is associated with a poor prognosis in NSCLC, particularly in lung ADCC. p-JAK1 is an independent poor prognostic factor and a potential target for early diagnosis. The combination of EGFR gene amplification and JAK1 activation may be a novel tool for prognosis prediction in patients with lung ADCC.

Acknowledgements

The authors of the present study are grateful to the pathologist from the Department of Pathology, Sichuan Provincial People's Hospital for assisting in the collection of the tissue samples. The present study was supported by grants from the National Natural Foundation of China (grant no. 81201851), Support Program of Science & Technology Department of Sichuan Province (grant no. 2014SZ0231) and Doctoral Foundation of Sichuan Provincial People's Hospital (grant no. 30305030563).

References

1 

Peters S, Adjei AA, Gridelli C, Reck M, Kerr K and Felip E: ESMO Guidelines Working Group: Metastatic non-small-cell lung cancer (NSCLC): ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 23:(Suppl 7). vii56–vii64. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Liu D, Huang Y, Chen B, Zeng J, Guo N, Zhang S, Liu L, Xu H, Mo X and Li W: Activation of mammalian target of rapamycin pathway confers adverse outcome in nonsmall cell lung carcinoma. Cancer. 117:3763–3773. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Rawlings JS, Rosler KM and Harrison DA: The JAK/STAT signaling pathway. J Cell Sci. 117:1281–1283. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Constantinescu SN, Leroy E, Gryshkova V, Pecquet C and Dusa A: Activating Janus kinase pseudokinase domain mutations in myeloproliferative and other blood cancers. Biochem Soc Trans. 41:1048–1054. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Bergmann AK, Schneppenheim S, Seifert M, Betts MJ, Haake A, Lopez C, Maria Murga, Penas E, Vater I, Jayne S, Dyer MJ, et al: Recurrent mutation of JAK3 in T-cell prolymphocytic leukemia. Genes Chromosomes Cancer. 53:309–316. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Wang SW and Sun YM: The IL-6/JAK/STAT3 pathway: Potential therapeutic strategies in treating colorectal cancer (Review). Int J Oncol. 44:1032–1040. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Gurbuz V, Konac E, Varol N, Yilmaz A, Gurocak S, Menevse S and Sozen S: Effects of AG490 and S3I-201 on regulation of the JAK/STAT3 signaling pathway in relation to angiogenesis in TRAIL-resistant prostate cancer cells. Oncol Lett. 7:755–763. 2014.PubMed/NCBI

9 

Schindler C, Levy DE and Decker T: JAK-STAT signaling: From interferons to cytokines. J Biol Chem. 282:20059–20063. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Xie H, Bae H, Noh J, Eun JW, Kim JK, Jung KH, Ryu JC, Ahn YM, Kim SY, Lee SH, et al: Mutational analysis of JAK1 gene in human hepatocellular carcinoma. Neoplasma. 56:136–140. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Jeong EG, Kim MS, Nam HK, Min CK, Lee S, Chung YJ, Yoo NJ and Lee SH: Somatic mutations of JAK1 and JAK3 in acute leukemias and solid cancers. Clin Cancer Res. 14:3716–3721. 2008. View Article : Google Scholar : PubMed/NCBI

12 

You Z, Xu D, Ji J, Guo W, Zhu W and He J: JAK/STAT signal pathway activation promotes progression and survival of human oesophageal squamous cell carcinoma. Clin Transl Oncol. 14:143–149. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Liu D, Huang Y, Zeng J, Chen B, Huang N, Guo N, Liu L, Xu H, Mo X and Li W: Down-regulation of JAK1 by RNA interference inhibits growth of the lung cancer cell line A549 and interferes with the PI3K/mTOR pathway. J Cancer Res Clin Oncol. 137:1629–1640. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Hirsch FR, Varella-Garcia M, McCoy J, West H, Xavier AC, Gumerlock P, Bunn PA Jr, Franklin WA, Crowley J and Gandara DR: Southwest Oncology Group: Increased epidermal growth factor receptor gene copy number detected by fluorescence in situ hybridization associates with increased sensitivity to gefitinib in patients with bronchioloalveolar carcinoma subtypes: A southwest oncology group study. J Clin Oncol. 23:6838–6845. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Lee Y, Shim HS, Park MS, Kim JH, Ha SJ, Kim SH and Cho BC: High EGFR gene copy number and skin rash as predictive markers for EGFR tyrosine kinase inhibitors in patients with advanced squamous cell lung carcinoma. Clin Cancer Res. 18:1760–1768. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Shan L, Wang Z, Guo L, Sun H, Qiu T, Ling Y, Li W, Li L, Liu X, Zheng B, et al: Concurrence of EGFR amplification and sensitizing mutations indicate a better survival benefit from EGFR-TKI therapy in lung adenocarcinoma patients. Lung Cancer. 89:337–342. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Koh Y, Jang B, Jeon YK, Kim TM, Lee SH, Kim DW, Chung DH, Kim YT, Kim YW and Heo DS: EGFR gene copy number gain is related to high tumor SUV and frequent relapse after adjuvant chemotherapy in resected lung adenocarcinoma. Jpn J Clin Oncol. 41:548–554. 2011. View Article : Google Scholar : PubMed/NCBI

18 

UICC and IUAC: TNM classification of malignant tumours. Wiley; New York, NY: 2002

19 

Travis W, Brambilla E, Muller-Hermlink H and Harris C: World Health Organization classification of tumoursPathology and genetics of tumours of the lung, pleura, thymus and heart. IARC Press; Lyon: 2004

20 

NCCN: NCCN Clinical practice guideline in oncology-non-small cell lung cancer guideline 2004. http://www.nccn.org2004.

21 

Dacic S, Flanagan M, Cieply K, Ramalingam S, Luketich J, Belani C and Yousem SA: Significance of EGFR protein expression and gene amplification in non-small cell lung carcinoma. Am J Clin Pathol. 125:860–865. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Varella-Garcia M: Stratification of non-small cell lung cancer patients for therapy with epidermal growth factor receptor inhibitors: The EGFR fluorescence in situ hybridization assay. Diagn Pathol. 1:192006. View Article : Google Scholar : PubMed/NCBI

23 

Murakami T, Takigawa N, Ninomiya T, Ochi N, Yasugi M, Honda Y, Kubo T, Ichihara E, Hotta K, Tanimoto M and Kiura K: Effect of AZD1480 in an epidermal growth factor receptor-driven lung cancer model. Lung Cancer. 83:30–36. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Hedvat M, Huszar D, Herrmann A, Gozgit JM, Schroeder A, Sheehy A, Buettner R, Proia D, Kowolik CM, Xin H, et al: The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell. 16:487–497. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Torres AF, Nogueira C, Magalhaes J, Costa IS, Aragao A, Gomes Neto A, Martins F and Tavora F: Expression of EGFR and molecules downstream to PI3K/Akt, Raf-1-MEK-1-MAP (Erk1/2), and JAK (STAT3) pathways in invasive lung adenocarcinomas resected at a single institution. Anal Cell Pathol (Amst). 2014:3529252014.PubMed/NCBI

26 

Cordero JB, Stefanatos RK, Myant K, Vidal M and Sansom OJ: Non-autonomous crosstalk between the Jak/Stat and Egfr pathways mediates Apc1-driven intestinal stem cell hyperplasia in the Drosophila adult midgut. Development. 139:4524–4535. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Huang Y, Li X, Jiang J and Frank SJ: Prolactin modulates phosphorylation, signaling and trafficking of epidermal growth factor receptor in human T47D breast cancer cells. Oncogene. 25:7565–7576. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2017
Volume 14 Issue 4

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
Liu D, Huang Y, Zhang L, Liang DN and Li L: Activation of Janus kinase 1 confers poor prognosis in patients with non‑small cell lung cancer. Oncol Lett 14: 3959-3966, 2017
APA
Liu, D., Huang, Y., Zhang, L., Liang, D., & Li, L. (2017). Activation of Janus kinase 1 confers poor prognosis in patients with non‑small cell lung cancer. Oncology Letters, 14, 3959-3966. https://doi.org/10.3892/ol.2017.6690
MLA
Liu, D., Huang, Y., Zhang, L., Liang, D., Li, L."Activation of Janus kinase 1 confers poor prognosis in patients with non‑small cell lung cancer". Oncology Letters 14.4 (2017): 3959-3966.
Chicago
Liu, D., Huang, Y., Zhang, L., Liang, D., Li, L."Activation of Janus kinase 1 confers poor prognosis in patients with non‑small cell lung cancer". Oncology Letters 14, no. 4 (2017): 3959-3966. https://doi.org/10.3892/ol.2017.6690